201
|
Tahghighi A, Seyedhashemi E, Mohammadi J, Moradi A, Esmaeili A, Pornour M, Jafarifar K, Ganji SM. Epigenetic marvels: exploring the landscape of colorectal cancer treatment through cutting-edge epigenetic-based drug strategies. Clin Epigenetics 2025; 17:34. [PMID: 39987205 PMCID: PMC11847397 DOI: 10.1186/s13148-025-01844-w] [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: 09/01/2024] [Accepted: 02/14/2025] [Indexed: 02/24/2025] Open
Abstract
Epigenetics is currently considered the investigation of inheritable changes in gene expression that do not rely on DNA sequence alteration. Significant epigenetic procedures are involved, such as DNA methylations, histone modifications, and non-coding RNA actions. It is confirmed through several investigations that epigenetic changes are associated with the formation, development, and metastasis of various cancers, such as colorectal cancer (CRC). The difference between epigenetic changes and genetic mutations is that the former could be reversed or prevented; therefore, cancer treatment and prevention could be achieved by restoring abnormal epigenetic events within the neoplastic cells. These treatments, consequently, cause the anti-tumour effects augmentation, drug resistance reduction, and host immune response stimulation. In this article, we begin our survey by exploring basic epigenetic mechanisms to understand epigenetic tools and strategies for treating colorectal cancer in monotherapy and combination with chemotherapy or immunotherapy.
Collapse
Affiliation(s)
- Azar Tahghighi
- Medicinal Chemistry Laboratory, Clinical Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Effat Seyedhashemi
- Department of Molecular Medicine, Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Shahrak-E Pajoohesh, Km 15, P.O. Box 14965/161, Tehran, Iran
| | - Javad Mohammadi
- Department of Molecular Medicine, Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Shahrak-E Pajoohesh, Km 15, P.O. Box 14965/161, Tehran, Iran
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Arash Moradi
- Department of Molecular Medicine, Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Shahrak-E Pajoohesh, Km 15, P.O. Box 14965/161, Tehran, Iran
| | - Aria Esmaeili
- Department of Molecular Medicine, Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Shahrak-E Pajoohesh, Km 15, P.O. Box 14965/161, Tehran, Iran
| | - Majid Pornour
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD, USA
| | - Kimia Jafarifar
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Shahla Mohammad Ganji
- Department of Molecular Medicine, Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Shahrak-E Pajoohesh, Km 15, P.O. Box 14965/161, Tehran, Iran.
| |
Collapse
|
202
|
Carranza FG, Waldrup B, Jin Y, Amzaleg Y, Postel M, Craig DW, Carpten JD, Salhia B, Hernandez D, Gutierrez N, Ricker CN, Culver JO, Chavez CE, Stern MC, Baezconde-Garbanati L, Lenz HJ, Velazquez-Villarreal E. Assessment of MYC Gene and WNT Pathway Alterations in Early-Onset Colorectal Cancer Among Hispanic/Latino Patients Using Integrated Multi-Omics Approaches. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.12.05.24318588. [PMID: 40034762 PMCID: PMC11875251 DOI: 10.1101/2024.12.05.24318588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Colorectal cancer (CRC) has increased at an alarming rate amongst younger (< 50 years) individuals. Such early-onset colorectal cancer (EOCRC) has been particularly notable within the Hispanic/Latino population. Yet, this population has not been sufficiently profiled in terms of two critical elements of CRC -- the MYC proto-oncogene and WNT signaling pathway. Here, we performed a comprehensive multi-omics analysis on 30 early-onset and 37 late-onset CRC (≥ 50 years) samples from Hispanic/Latino patients. Our analysis included DNA exome sequencing for somatic mutations, somatic copy number alterations, and global and local genetic similarity. Using RNA sequencing, we also assessed differential gene expression, cellular pathways, and gene fusions. We then compared our findings from early-onset Hispanic/Latino patient samples with publicly available data from Non-Hispanic White cohorts. Across all early-onset patients, which had a median 1000 Genomes Project Peruvian-in-Lima-like (1KG-PEL-like) genetic similarity proportion of 60%, we identified 41 WNT pathway genes with significant mutations. Six important examples were APC, TCF7L2, DKK1, DKK2, FZD10, and LRP5. Notably, patients with mutations in DKK1 and DKK2 had the highest 1KG-PEL-like proportion (79%). When we compared the Hispanic/Latino cohort to the Non-Hispanic White cohorts, four of these key genes -- DKK1, DKK2, FZD10, and LRP5 -- were significant in both risk association analyses and differential gene expression. Interestingly, early-onset tumors (vs. late-onset) exhibited distinct somatic copy number alterations and gene expression profiles; the differences included MYC and drug-targetable WNT pathway genes. We also identified a novel WNT gene fusion, RSPO3, in early-onset tumors; it was associated with enhanced WNT signaling. This integrative analysis underscores the distinct molecular features of EOCRC cancer in the Hispanic/Latino population; reveals potential avenues for tailored precision medicine therapies; and emphasizes the importance of multi-omics approaches in studying colorectal carcinogenesis. We expect this data to help contribute towards reducing cancer health disparities. Significance This study offers multi-omics profiling analysis of early-onset colorectal cancer (EOCRC) in an underserved community, explores the implications of MYC gene and WNT pathway alterations, and provides critical insights into cancer health disparities.
Collapse
Affiliation(s)
- F G Carranza
- City of Hope, Beckman Research Institute, Department of Integrative Translational Sciences, Duarte, CA
| | - B Waldrup
- City of Hope, Beckman Research Institute, Department of Integrative Translational Sciences, Duarte, CA
| | - Y Jin
- City of Hope, Beckman Research Institute, Department of Integrative Translational Sciences, Duarte, CA
| | - Y Amzaleg
- City of Hope, Beckman Research Institute, Department of Integrative Translational Sciences, Duarte, CA
| | - M Postel
- University of Southern California, Keck School of Medicine of USC, Department of Translational Genomics, Los Angeles, CA
| | - D W Craig
- City of Hope, Beckman Research Institute, Department of Integrative Translational Sciences, Duarte, CA
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - J D Carpten
- City of Hope, Beckman Research Institute, Department of Integrative Translational Sciences, Duarte, CA
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - B Salhia
- University of Southern California, Keck School of Medicine of USC, Department of Translational Genomics, Los Angeles, CA
- University of Southern California, USC Norris Comprehensive Cancer Center, Los Angeles, CA
| | - D Hernandez
- University of Southern California, Keck School of Medicine of USC, Division of Medical Oncology, Los Angeles, CA
| | - N Gutierrez
- University of Southern California, Keck School of Medicine of USC, Division of Medical Oncology, Los Angeles, CA
| | - C N Ricker
- University of Southern California, USC Norris Comprehensive Cancer Center, Los Angeles, CA
- University of Southern California, Keck School of Medicine of USC, Division of Medical Oncology, Los Angeles, CA
- Los Angeles General Medical Center, Los Angeles, CA
| | - J O Culver
- University of Southern California, USC Norris Comprehensive Cancer Center, Los Angeles, CA
- University of Southern California, Keck School of Medicine of USC, Division of Medical Oncology, Los Angeles, CA
| | - C E Chavez
- University of Southern California, USC Norris Comprehensive Cancer Center, Los Angeles, CA
| | - M C Stern
- University of Southern California, USC Norris Comprehensive Cancer Center, Los Angeles, CA
- University of Southern California, Keck School of Medicine of USC, Department of Population and Public Health Sciences, Los Angeles, CA
| | - L Baezconde-Garbanati
- University of Southern California, USC Norris Comprehensive Cancer Center, Los Angeles, CA
- University of Southern California, Keck School of Medicine of USC, Department of Population and Public Health Sciences, Los Angeles, CA
| | - H J Lenz
- University of Southern California, USC Norris Comprehensive Cancer Center, Los Angeles, CA
- University of Southern California, Keck School of Medicine of USC, Division of Medical Oncology, Los Angeles, CA
| | - E Velazquez-Villarreal
- City of Hope, Beckman Research Institute, Department of Integrative Translational Sciences, Duarte, CA
- City of Hope Comprehensive Cancer Center, Duarte, CA
| |
Collapse
|
203
|
Ghabra S, Chang D, Sugarbaker PH. Preoperative tumor marker elevations in colorectal cancer patients with peritoneal metastases should be used to help select patients for cytoreductive surgery. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109720. [PMID: 40023022 DOI: 10.1016/j.ejso.2025.109720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/04/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Tumor markers are used routinely for surveillance in patients treated for colorectal cancer (CRC). However, the prognostic implications of elevated preoperative tumor markers in patients treated for CRC and peritoneal metastases (PM) has not been well defined. The utility of carcinoembryonic antigen (CEA), carbohydrate antigen 19-9 (CA19-9) and cancer antigen 125 (CA125) to predict outcome for these patients is reported. METHODS Clinical and histologic features plus preoperative tumor markers were recorded within 1 week prior to CRS. Impact on overall survival of these factors was analyzed by univariate and multivariate analysis. RESULTS Two hundred and four patients were in our database and 140 patients (75.3 %) had at least a single elevated preoperative tumor marker. In an analysis of clinical and histologic parameters preoperatively, a poorly differentiated tumor, signet ring morphology, a peritoneal cancer index (PCI) of ≥16 and an incomplete cytoreduction had a negative impact on median survival. In a multivariate analysis of clinical and histologic features together with tumor markers, an elevated CA19-9 and CA125 was independently associated with reduced overall survival (HR 2.7, p < 0.0001 and HR 2.2, p = 0.005), respectively. Quantitative assessment of CEA (HR 0.5, p = 0.0094) and CA19-9 (HR 4.9, p < 0.001) greater than x10 ULN showed reduced survival. CONCLUSION Preoperative assessment of symptoms and histopathology, PCI and a complete CRS combined with tumor markers CEA, CA19-9 and CA125 are independent prognostic indicators for selection by the multidisciplinary team of CRC PM patients for CRS and HIPEC. All three tumor markers are needed for a meaningful assessment.
Collapse
Affiliation(s)
- Shadin Ghabra
- Department of Surgery, MedStar Washington Hospital Center, Washington, DC, USA
| | | | - Paul H Sugarbaker
- Department of Surgery, MedStar Washington Hospital Center, Washington, DC, USA.
| |
Collapse
|
204
|
Yang G, Qian B, He L, Zhang C, Wang J, Qian X, Wang Y. Application prospects of ferroptosis in colorectal cancer. Cancer Cell Int 2025; 25:59. [PMID: 39984914 PMCID: PMC11846473 DOI: 10.1186/s12935-025-03641-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 01/07/2025] [Indexed: 02/23/2025] Open
Abstract
Colorectal cancer (CRC) is a serious threat to human health with the third morbidity and the second cancer-related mortality worldwide. It is urgent to explore more effective strategy for CRC because of the acquired treatment resistance from the non-surgical conventional therapies, including radiation, chemotherapy, targeted therapy and immunotherapy. Ferroptosis is a novel form of programmed cell death characterized by iron-dependent lipid peroxidation species (ROS) accumulation and has been identified as a promising target for cancer treatment, especially for those with treatment resistance. In this review, we mainly summarize the recent studies on the influence and regulation of ferroptosis by which (including gut microbiota) modulating the metabolism of iron, amino acid and lipid. Thus this analysis may provide potential targets for inducing CRC ferroptosis and shed lights on the future application of ferroptosis in CRC.
Collapse
Affiliation(s)
- Gen Yang
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Boning Qian
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Liya He
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Chi Zhang
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Jianqiang Wang
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xinlai Qian
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Henan Provincial Key Laboratory of Molecular Oncologic Pathology, Xinxiang, Henan, China.
| | - Yongxia Wang
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
- Henan Provincial Key Laboratory of Molecular Oncologic Pathology, Xinxiang, Henan, China.
| |
Collapse
|
205
|
Huang M, Li J, Huang W, Zhou Y, Cai L, Liu M. The effectiveness of Evodia rutaecarpa hot compress on the recovery of gastrointestinal function after laparoscopic surgery for colorectal cancer: A propensity score-matched retrospective cohort study. PLoS One 2025; 20:e0303951. [PMID: 39977411 PMCID: PMC11841865 DOI: 10.1371/journal.pone.0303951] [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: 05/05/2024] [Accepted: 11/05/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Although the use of hot compresses with the herbal medicine Evodia rutaecarpa (ER) as a complementary and alternative therapy to promote recovery of postoperative gastrointestinal function is gradually increasing in clinical practice, there is still a lack of relevant empirical studies. Particularly, the role of ER hot compress therapy on gastrointestinal recovery post-laparoscopic surgery for colorectal cancer has not been well investigated. The purpose of this study is to evaluate the efficacy and applicability of ER hot compress therapy for the recovery of postoperative gastrointestinal function. METHODS This is a retrospective cohort study. Patients were divided into two cohorts, the ER group and the non-ER group. Propensity score matching(PSM) was introduced to limit confounding, and independent samples t-tests, non-parametric tests, or Chi-squared tests were used to compare these two cohorts. RESULTS A total of 454 patients were included, with 267 (59%) receiving ER hot compress therapy and 187 (41%) not. After 1:1 PSM, 320 patients were analyzed (160 in each group). Compared to the ER group, patients in the non-ER group had shorter times to return to a semi-liquid diet (p = 0.030) and hospital stay (p<0.001), as well as lower hospital costs (p<0.001). Subgroup analyses revealed no statistically significant differences in the length of hospital stay, hospital costs, postoperative time to return to full-liquid diet, or time to return to semi-liquid diet among stage I and II tumor patients. However, except for hospital costs, the means and standard deviations of the other indicators were generally lower in the ER group. Complication incidence showed no significant difference between the two cohorts before and after PSM. CONCLUSIONS The use of ER hot packs after laparoscopic surgery in patients with colorectal cancer has a non-significant effect on the recovery of the gastrointestinal function and, given the results of the study, it is likely that patients with early-stage tumors may benefit more. Therefore, healthcare providers need to consider the individualization, practicality, and economics of treatment options.
Collapse
Affiliation(s)
- Miaoxin Huang
- Faculty of Health Sciences and Sports, Macao Polytechnic University, Macao SAR, China
| | - Junmiao Li
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wei Huang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yuling Zhou
- The Third Affiliated Hospital of Sun Yat sen University, Guangzhou, Guangdong, China
| | - Lei Cai
- The Third Affiliated Hospital of Sun Yat sen University, Guangzhou, Guangdong, China
| | - Ming Liu
- Peking University Health Science Center—Macao Polytechnic University Nursing Academy, Macao Polytechnic University, Macao SAR, China
| |
Collapse
|
206
|
Iwamoto Y, Salmon B, Yoshioka Y, Kojima R, Krull A, Ota S. High throughput analysis of rare nanoparticles with deep-enhanced sensitivity via unsupervised denoising. Nat Commun 2025; 16:1728. [PMID: 39979247 PMCID: PMC11842628 DOI: 10.1038/s41467-025-56812-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 01/31/2025] [Indexed: 02/22/2025] Open
Abstract
The large-scale multiparametric analysis of individual nanoparticles is increasingly vital in the diverse fields of biology, medicine, and materials science. However, the current methods struggle with the tradeoff between measurement scalability and sensitivity, especially when identifying rare nanoparticles in heterogeneous mixtures. By developing and combining an unsupervised deep learning-based denoising method and an optofluidic device tuned for nanoparticle detection, we realize a nanoparticle analyzer that simultaneously achieves high scalability, throughput, and sensitivity levels; we name this approach "Deep Nanometry" (DNM). DNM detects polystyrene beads with a detection of limit of 30 nm at a throughput of over 100,000 events/second. The sensitive and scalable DNM directly detects rare target extracellular vesicles (EVs) in non-purified serum, making up as little as 0.002% of the 1,214,392 total particles. Moreover, DNM accurately and sufficiently counts diagnostic marker EVs present in only 0.93% and 0.17% of particle detections in sera of colorectal cancer patients and healthy controls, demonstrating its potential application to the early detection of colorectal cancer.
Collapse
Affiliation(s)
- Yuichiro Iwamoto
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro 4-6-1, Shibuya, Tokyo, Japan
| | - Benjamin Salmon
- School of Computer Science, University of Birmingham, Birmingham, UK
| | - Yusuke Yoshioka
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Nishishinjuku 6-7-1, Shinjuku, Tokyo, Japan
| | - Ryosuke Kojima
- Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo, Tokyo, Japan
| | - Alexander Krull
- School of Computer Science, University of Birmingham, Birmingham, UK.
| | - Sadao Ota
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro 4-6-1, Shibuya, Tokyo, Japan.
| |
Collapse
|
207
|
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
|
208
|
He M, Gu R, Huang X, Zhao A, Tian S, Zheng Y. Global burden of colorectal cancer attributable to metabolic risks from 1990 to 2021, with projections of mortality to 2035. Int J Colorectal Dis 2025; 40:46. [PMID: 39969585 PMCID: PMC11839689 DOI: 10.1007/s00384-025-04817-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND The global burden of colorectal cancer (CRC) attributable to metabolic risk factors is increasing. It is crucial to analyze the global epidemiological patterns of CRC attributable to metabolic risk factors and predict future trends. METHODS Detailed data on CRC mortality and disability-adjusted life years (DALYs) attributable to metabolic risk factors were extracted for this study using data from the Global Burden of Diseases (GBD) 2021 study to assess the burden of CRC from 1990 to 2021 by global, regional, national, and sociodemographic index (SDI) regions and quantify the time trend using the estimated annual percentage change (EAPC). Bayesian age-period-cohort (BAPC) models projected the global mortality from 2022 to 2035. RESULTS The global number of CRC deaths due to metabolic risk factors increased from 70,916 in 1990 to 172,993 in 2021, a 2.4-fold increase. CRC mortality and DALYs attributable to high body mass index (BMI) and fasting plasma glucose (FPG) increased significantly at the global level from 1990 to 2021. In 2021, the GBD regions with the highest CRC age-standardized mortality rate (ASMR) due to high BMI and high FPG were in Central Europe. The ASMR of CRC attributable to high BMI and high FPG among males is expected to increase from 2022 to 2035. CONCLUSION CRC mortality and DALYs attributable to metabolic factors are increasing. Reducing the burden of CRC due to high BMI and FPG levels is critically needed.
Collapse
Affiliation(s)
- Maolang He
- Department of Gastroenterology, The First Affiliated Hospital of Shihezi University, Shihezi, 832008, China
| | - Ruru Gu
- Department of Gastroenterology, The First Affiliated Hospital of Shihezi University, Shihezi, 832008, China
| | - Xin Huang
- School of Medicine, Shihezi University, Shihezi, 832002, China
| | - Aifang Zhao
- School of Medicine, Shihezi University, Shihezi, 832002, China
| | - Shuxin Tian
- Department of Gastroenterology, The First Affiliated Hospital of Shihezi University, Shihezi, 832008, China.
| | - Yong Zheng
- Department of Gastroenterology, The First Affiliated Hospital of Shihezi University, Shihezi, 832008, China.
| |
Collapse
|
209
|
Taghizadeh-Teymorloei M, Jafarlou V, Matin S, Raeisi M, Roosta Y, Mansouri-Derakhshani S, Feizi AAH, Karimi A. Clinical implications of Alu-based cell-free DNA and serum onco-piRNA monitoring in colorectal cancer management. Clin Transl Oncol 2025:10.1007/s12094-025-03863-8. [PMID: 39969763 DOI: 10.1007/s12094-025-03863-8] [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: 12/17/2024] [Accepted: 01/28/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) remains a significant global health challenge, characterized by high morbidity and mortality rates. This study explores the potential of Alu-based cell-free DNA (cfDNA) and specific PIWI-interacting RNAs (piRNAs) as innovative biomarkers for monitoring treatment responses in CRC patients. METHODS We analyzed plasma samples from 70 CRC patients, equally divided between those undergoing chemotherapy and surgical interventions. RESULTS Our findings reveal that certain piRNAs, particularly piRNA-823, piRNA-54265, and piRNA-1245, exhibit significant prognostic value, with notable expression changes observed in the chemotherapy group compared to the surgery group. Furthermore, the levels of ALU-based cfDNA fragments showed a marked decrease post-chemotherapy, suggesting their utility in assessing therapeutic efficacy. CONCLUSIONS This research underscores the importance of integrating these molecular tools particularly piRNA-823 and ALU-based cfDNA into clinical practice, potentially enhancing the management strategies for CRC patients and improving their outcomes.
Collapse
Affiliation(s)
- Mohammad Taghizadeh-Teymorloei
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht St., Tabriz, 5166614756, East Azerbaijan, Iran
| | - Vahid Jafarlou
- Cancer Institute of Imam Khomeini Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Somaieh Matin
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mortaza Raeisi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Roosta
- Department of Internal Medicine, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Sima Mansouri-Derakhshani
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Genetics, Tabriz, Iran
| | - Abbas Ali Hosseinpour Feizi
- Hematology-Oncology Research Center, Tabriz University of Medical Sciences, Tabriz Children's Hospital, Tabriz, Iran
| | - Abbas Karimi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht St., Tabriz, 5166614756, East Azerbaijan, Iran.
| |
Collapse
|
210
|
Al-Alam ODCM, Alves RJV, Fae AL, Cardoso CN, Lava JR, Castro JP, Signori J, Claudino LG, Wandscheer TBC, Villarinho VV, Yao APMS, Bica CG. Clinical and demographic analysis of patients with colorectal cancer screened at a reference hospital in Southern Brazil: comparative study based on age (Retrospective cohort study). BMC Gastroenterol 2025; 25:91. [PMID: 39966718 PMCID: PMC11837471 DOI: 10.1186/s12876-025-03607-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/10/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a prevalent and lethal malignancy. This study examines differences in clinical and prognostic characteristics of CRC between patients under 50 years and those aged 50 and above, treated at a reference hospital in Southern Brazil. METHODS A retrospective cohort study was conducted, analyzing data from the Cancer Hospital Registry in Porto Alegre, RS. Patients diagnosed with colon or rectal adenocarcinoma between January 2013 and December 2017 were included. Variables analyzed included family history, alcoholism, smoking status, clinical staging, tumor laterality, clinical presentation, CEA levels at diagnosis, adjuvant chemotherapy, and neutrophil-to-lymphocyte ratio. RESULTS The study cohort included 1,121 patients, with 85% aged 50 years or older, and 15% younger than 50 years. Significant differences were observed in smoking status and clinical presentation, with younger patients presenting with a higher prevalence of obstruction/subocclusion at diagnosis, which may reflect differences in clinical presentation due to delayed diagnosis. CONCLUSIONS Age and lifestyle factors, particularly smoking, significantly influenced the clinical presentation and management of colorectal cancer. These findings underscore the importance of targeted prevention strategies and personalized treatment approaches for younger CRC patients.
Collapse
Affiliation(s)
- Otavio de Carvalho Modaffar Al-Alam
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil.
- Postdoctoral Research Fellow, Johns Hopkins University, Baltimore, USA.
| | - Rafael Jose Vargas Alves
- Department of Internal Medicine, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
- Clinical Oncology Department, Hospital Santa Rita, Santa Casa Hospital, Porto Alegre, Brazil
| | - Adele Lanziani Fae
- Undergraduate Medicine Program, Lutheran University of Brazil (ULBRA), Canoas, Brazil
| | - Camile Neves Cardoso
- Undergraduate Medicine Program, Lutheran University of Brazil (ULBRA), Canoas, Brazil
| | - Jamille Rizzardi Lava
- Clinical Oncology Department, Hospital Santa Rita, Santa Casa Hospital, Porto Alegre, Brazil
| | | | - Julia Signori
- Clinical Oncology Department, Hospital Santa Rita, Santa Casa Hospital, Porto Alegre, Brazil
| | | | | | | | | | - Claudia Giuliano Bica
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
- Department of Basic Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| |
Collapse
|
211
|
Passardi A, Gibbons D, Scanu AM, Dhar SS. Editorial: Molecular mechanisms and targeted therapies for colorectal cancer vol II. Front Oncol 2025; 15:1566681. [PMID: 40040718 PMCID: PMC11876110 DOI: 10.3389/fonc.2025.1566681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 02/05/2025] [Indexed: 03/06/2025] Open
Affiliation(s)
- Alessandro Passardi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - David Gibbons
- Department of Histopathology, St. Vincent’s University Hospital, Dublin, Ireland
- The School of Medicine and Health Sciences, University College, Dublin, Ireland
| | - Antonio Mario Scanu
- Department of Medicine, Surgery and Pharmacy, Unit of General Surgery 2, University of Sassari, Sassari, Italy
| | - Shilpa S Dhar
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
212
|
Wang W, Dai Z, Ge C, Zhou X, Zhan Y, Chen C. Comprehensive analysis of basement membrane-related genes showed that NELL2 is a new therapeutic target for colorectal cancer. Discov Oncol 2025; 16:195. [PMID: 39961875 PMCID: PMC11832829 DOI: 10.1007/s12672-025-01979-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND The basement membrane (BM), an omnipresent extracellular matrix, plays a pivotal role as a physiological element in the process of tumor metastasis. However, given the heterogeneity of colorectal cancer (CRC), prognosis is challengingly predictive. Therefore, we aim to construct a prognostic model using BM-associated genes to assess patient prognosis and clinical drug treatment effects. METHOD The Non-negative Matrix Factorization (NMF) algorithm leverages the characteristics or categories of matrix rows and columns to achieve BMAG molecular classification and further develop a model for predicting patient prognosis. ssGSEA quantified the relatively abundance of 13 immune functionalities and 16 immune cell typologies. To predict the efficacy of immunotherapy, a comprehensive investigation was conducted on the correlations between riskScores and key factors such as TME, immune checkpoints, and MMR-related genes. The CCK8 method, plate cloning method and Cell Apoptosis Assessment were used to evaluate the ability of NELL2 to affect the proliferation. RESULT We developed a powerful riskScore to predict colorectal cancer prognosis and effectively differentiate the tumor microenvironment. In clinical practice, this riskScore can also be utilized to further assess patient prognosis, thereby facilitating personalized treatment strategies. In addition, downregulation of NELL2 expression inhibits CRC cell proliferation. CONCLUSION In summary, we constructed a novel riskScore using BMAG for predicting prognosis in patients with CRC and explored the efficacy of this riskScore in predicting patient response to clinical drug therapy. Most importantly, we have identified the oncogenic role of NELL2 in CRC. By inhibiting NELL2, we can further suppress the initiation and progression of CRC.
Collapse
Affiliation(s)
- Weiguo Wang
- Department of Interventional and Vascular Surgery, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Zhengxing Dai
- Department of Interventional and Vascular Surgery, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Chen Ge
- Department of General Surgery, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Xitian Zhou
- Department of Interventional and Vascular Surgery, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Yi Zhan
- Department of Interventional and Vascular Surgery, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Chaobo Chen
- Department of General Surgery, Xishan People's Hospital of Wuxi City, Wuxi, China.
| |
Collapse
|
213
|
Ghidini M, Hahne JC, Senti C, Heide T, Proszek PZ, Shaikh R, Carter P, Hubank M, Trevisani F, Garrone O, Cappelletti MR, Generali D, Cattaneo M, Gnocchi N, Donati G, Gobbi A, Grizzi G, Lampis A, Elghadi R, Tanzi G, Tomasello G, Ratti M, Pinato DJ, Fassan M, Vlachogiannis G, Sottoriva A, Cortellini A, Passalacqua R, Valeri N. Circulating Tumor DNA Dynamics and Clinical Outcome in Metastatic Colorectal Cancer Patients Undergoing Front-Line Chemotherapy. Clin Cancer Res 2025; 31:707-718. [PMID: 39688961 DOI: 10.1158/1078-0432.ccr-24-0924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 08/01/2024] [Accepted: 12/13/2024] [Indexed: 12/19/2024]
Abstract
PURPOSE We tested whether circulating tumor DNA (ctDNA) changes may be used to assess early response and clinical outcomes in patients with metastatic colorectal cancer (mCRC) undergoing first-line systemic anticancer therapy (SACT). EXPERIMENTAL DESIGN Eight hundred sixty-two plasma samples were collected 4-weekly from baseline (BL) until disease progression in patients with mCRC receiving first-line SACT. ctDNA was tested using tissue-agnostic next-generation sequencing panels. ctDNA normalization was defined as ≥99% clearance after 1 month of therapy (Mo1) in the three variants with the highest allele frequency in BL ctDNA. RESULTS Eighty-three paired samples from 75 patients were available for analysis. Twelve pairs (14.4%) showed no variants in either BL or Mo1. In the remaining 71 comparisons (65 patients), 37 (52.1%) showed ctDNA normalization at Mo1. Patients who cleared ctDNA had significantly longer overall (45.6 months) and progression-free survival (13.9 months) compared with nonnormalized patients [overall survival = 22.6 months (log-rank P = 0.01) and progression-free survival = 10.7 months (log-rank P = 0.036), respectively]. In addition, a higher response rate was observed in patients with ctDNA clearance (72.9%) compared with nonnormalized cases (38.2%). Longitudinal sequencing of at least four time points in patients with a progression-free survival of >10 months showed emerging variants in 47.8% of cases; in all these patients, the trajectory of these new "outlier" variants seemed in stark contrast with the clinical-radiological course of disease and the trend in other mutations. CONCLUSIONS ctDNA clearance represents an early indicator of benefit from SACT in patients with mCRC; serial tracking of multiple variants is warranted to improve specificity and avoid misleading information due to the emergence of mutations of unknown clinical significance.
Collapse
Affiliation(s)
- Michele Ghidini
- Division of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Jens Claus Hahne
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- CRUK Experimental Cancer Centre and NIHR Biomedical Research Centre, University of Leicester, Leicester, United Kingdom
| | - Chiara Senti
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | | | - Paula Z Proszek
- Clinical Genomics Translational Research Laboratory, Royal Marsden Hospital, London, United Kingdom
| | - Ridwan Shaikh
- Clinical Genomics Translational Research Laboratory, Royal Marsden Hospital, London, United Kingdom
| | - Paul Carter
- Clinical Genomics Translational Research Laboratory, Royal Marsden Hospital, London, United Kingdom
| | - Mike Hubank
- Clinical Genomics Translational Research Laboratory, Royal Marsden Hospital, London, United Kingdom
| | - Francesco Trevisani
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Ornella Garrone
- Division of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Rosa Cappelletti
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, Cremona, Italy
| | - Daniele Generali
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, Cremona, Italy
| | - Monica Cattaneo
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - Nicoletta Gnocchi
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - Gianvito Donati
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - Angela Gobbi
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - Giulia Grizzi
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - Andrea Lampis
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Raghad Elghadi
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Giulia Tanzi
- Division of Pathology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - Gianluca Tomasello
- Division of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Margherita Ratti
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - David J Pinato
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
- Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Matteo Fassan
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
- Department of Medicine, Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Georgios Vlachogiannis
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Human Technopole, Milan, Italy
| | - Alessio Cortellini
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Rodolfo Passalacqua
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - Nicola Valeri
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| |
Collapse
|
214
|
Hong I, Kim S, Lee M, Han S, Kim HC, Chu CW, Kim SG, Kim MK, Kim CJ, Kang DH, Ahn TS, Baek MJ, Hussain M, Kwon HY, Jeong D. TM7SF2 as a Potential Biomarker in Colorectal Cancer: Implications for Metastasis. Curr Oncol 2025; 32:114. [PMID: 39996914 PMCID: PMC11854686 DOI: 10.3390/curroncol32020114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Colorectal cancer (CRC) is a commonly fatal cancer and ranks as the fourth most prevalent in men and third in women worldwide. While early-stage survival rates are high, they significantly decrease with recurrence and metastasis. Thus, the early detection and treatment of metastasis-related factors can significantly improve survival rates. In this study, the transmembrane 7 superfamily member 2 (TM7SF2) gene was validated as a biomarker for predicting metastasis in CRC. Immunohistochemical staining was performed on 236 CRC tissues, and the clinicopathological factors of patients with CRC were analyzed. This evaluation revealed that TM7SF2 expression is associated with the clinical stage. Kaplan-Meier analysis confirmed the relationship between the survival rate of CRC patients and TM7SF2 expression, showing a decrease in survival rate with TM7SF2 overexpression (log-rank, p < 0.001). TM7SF2 expression was also confirmed in two pairs of primary and metastatic cell lines (SW480 and SW620). TM7SF2 knockdown was executed using siRNAs in SW480 and SW620 cells, which exhibit high expression levels. The knockdown was verified using RT-PCR and immunoblotting. Functional studies investigated the effects of TM7SF2 on cell proliferation, migration, invasion, and colony formation, revealing that all these functions were suppressed in the CRC cell lines following TM7SF2 knockdown. Therefore, TM7SF2 shows promise as a biomarker for the prevention of colorectal cancer.
Collapse
Affiliation(s)
- Inpyo Hong
- Department of Pathology, College of Medicine, Soonchunhyang University, Dongnam-gu, Cheonan 31151, Republic of Korea; (I.H.); (S.K.); (M.L.); (S.H.)
| | - Sooyoun Kim
- Department of Pathology, College of Medicine, Soonchunhyang University, Dongnam-gu, Cheonan 31151, Republic of Korea; (I.H.); (S.K.); (M.L.); (S.H.)
| | - Minho Lee
- Department of Pathology, College of Medicine, Soonchunhyang University, Dongnam-gu, Cheonan 31151, Republic of Korea; (I.H.); (S.K.); (M.L.); (S.H.)
| | - Seoin Han
- Department of Pathology, College of Medicine, Soonchunhyang University, Dongnam-gu, Cheonan 31151, Republic of Korea; (I.H.); (S.K.); (M.L.); (S.H.)
| | - Hak Chun Kim
- Soonchunhyang Medical Science Research Institute, College of Medicine, Soonchunhyang University, Dongnam-gu, Cheonan 31151, Republic of Korea;
| | - Chong Woo Chu
- Changwon Hanmaeum Hospital, Uichang-gu, Changwon-si 51139, Republic of Korea; (C.W.C.); (S.G.K.); (M.K.K.); (C.J.K.)
| | - Seong Geun Kim
- Changwon Hanmaeum Hospital, Uichang-gu, Changwon-si 51139, Republic of Korea; (C.W.C.); (S.G.K.); (M.K.K.); (C.J.K.)
| | - Min Kyung Kim
- Changwon Hanmaeum Hospital, Uichang-gu, Changwon-si 51139, Republic of Korea; (C.W.C.); (S.G.K.); (M.K.K.); (C.J.K.)
| | - Chang Jin Kim
- Changwon Hanmaeum Hospital, Uichang-gu, Changwon-si 51139, Republic of Korea; (C.W.C.); (S.G.K.); (M.K.K.); (C.J.K.)
| | - Dong Hyun Kang
- Department of Surgery, College of Medicine, Soonchunhyang University, Dongnam-gu, Cheonan 31151, Republic of Korea; (D.H.K.); (T.S.A.); (M.J.B.)
| | - Tae Sung Ahn
- Department of Surgery, College of Medicine, Soonchunhyang University, Dongnam-gu, Cheonan 31151, Republic of Korea; (D.H.K.); (T.S.A.); (M.J.B.)
| | - Moo Jun Baek
- Department of Surgery, College of Medicine, Soonchunhyang University, Dongnam-gu, Cheonan 31151, Republic of Korea; (D.H.K.); (T.S.A.); (M.J.B.)
| | - Mudasir Hussain
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si 31151, Republic of Korea; (M.H.); (H.Y.K.)
| | - Hyog Young Kwon
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si 31151, Republic of Korea; (M.H.); (H.Y.K.)
| | - Dongjun Jeong
- Department of Pathology, College of Medicine, Soonchunhyang University, Dongnam-gu, Cheonan 31151, Republic of Korea; (I.H.); (S.K.); (M.L.); (S.H.)
| |
Collapse
|
215
|
Wang ZH, Zeng X, Huang W, Yang Y, Zhang S, Yang M, Liu H, Zhao F, Li A, Zhang Z, Liu J, Shi J. Bioactive nanomotor enabling efficient intestinal barrier penetration for colorectal cancer therapy. Nat Commun 2025; 16:1678. [PMID: 39956840 PMCID: PMC11830786 DOI: 10.1038/s41467-025-57045-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 02/10/2025] [Indexed: 02/18/2025] Open
Abstract
Oral drug delivery systems had natural potential for colorectal cancer drug therapy. While the drug delivery efficiency is severely hindered by the complex intestinal barriers, especially mucus and epithelium barriers, resulting in unsatisfactory therapeutic effects and limited clinical translation. In this work, a bioactive self-thermophoretic and gas dual-driven nanomotor is developed for colorectal cancer therapy through efficient intestinal mucus and epithelial barrier penetration. The nanomotor shows intestinal mucus barrier penetration and the paracellular pathway reversibly opening properties of intestinal epithelium barrier, increasing the delivery efficiency of cisplatin by 3.5 folds. Owing to the targeted delivery of cisplatin and the reduced side effects on normal intestinal tissues, the therapeutic efficiency of the nanomotor for colorectal cancer in vivo is as high as 98.6%. With autonomous and reversible intestinal barriers penetration property, the nanoplatform may innovate the current oral drug delivery.
Collapse
Affiliation(s)
- Zhi-Hao Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, 450001, China
| | - Xuejiao Zeng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Wanting Huang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Yanbo Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Shuhao Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Mingzhu Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Hua Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Fengqin Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Airong Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou, 450001, China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, 450001, China.
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou, 450001, China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, 450001, China.
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou, 450001, China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, 450001, China.
| |
Collapse
|
216
|
Chen HLR, Chong QD, Tay B, Zhou S, Wong EYT, Seow-En I, Tan KK, Wang Y, Seow A, Tan KWE, Tan BHI, Tan SH. Trends in Early-Onset Colorectal Cancer in Singapore: Epidemiological Study of a Multiethnic Population. JMIR Public Health Surveill 2025; 11:e62835. [PMID: 39725547 PMCID: PMC11888020 DOI: 10.2196/62835] [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: 06/09/2024] [Revised: 11/02/2024] [Accepted: 12/24/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) incidence and mortality in those aged 50 years and above have decreased over the past 2 decades. However, there is a rising incidence of CRC among individuals under 50 years of age, termed early-onset colorectal cancer (EOCRC). Patients with EOCRC are diagnosed at an advanced stage and may be in more psychosocial, emotional, and financial distress. OBJECTIVE Our study examined the epidemiological shifts in CRC in Singapore, a multiethnic country. METHODS CRCs diagnosed at age 20 years and above were identified from the Singapore Cancer Registry (SCR) from 1968 to 2019. Patient characteristics included gender, ethnicity, and age of CRC diagnosis. Population information was obtained from the Department of Statistics Singapore (SingStat). Age-specific incidence rates (ASRs) and age-standardized incidence rates (ASIRs) were calculated. The cohort was divided into 3 age groups: 20-49, 50-64, and ≥65 years. Temporal trends in incidence rates were modeled with joinpoint regression. Birth cohort models were fitted using the National Cancer Institute (NCI) age-period-cohort analysis tool. Cancer-specific survival analysis was performed with the Cox proportional hazards model. RESULTS In total, 53,044 CRCs were included, and 6183 (11.7%) adults aged 20-49 years were diagnosed with EOCRC. The ASR of EOCRC rose from 5 per 100,000 population in 1968 to 9 per 100,000 population in 1996 at 2.1% annually and rose to 10 per 100,000 population in 2019 at 0.64% annually. The ASR for CRC among adults aged 50-64 years rose at 3% annually from 1968 to 1987 and plateaued from 1987, while the ASR for adults aged 65 years and above rose at 4.1% annually from 1968 to 1989 and 1.3% annually from 1989 to 2003 but decreased from 2003 onwards at 1% annually. The ASR of early-onset rectal cancer increased significantly at 1.5% annually. There was a continued rise in the ASR of EOCRC among males (annual percentage change [APC] 1.5%) compared to females (APC 0.41%). Compared to the 1950-1954 reference birth cohort, the 1970-1984 birth cohort had a significantly higher incidence rate ratio (IRR) of 1.17-1.36 for rectal cancer, while there was no significant change for colon cancer in later cohorts. There were differences in CRC trends across the 3 ethnic groups: Malays had a rapid and persistent rise in the ASR of CRC across all age groups (APC 1.4%-3%), while among young Chinese, only the ASR of rectal cancer was increasing (APC 1.5%). Patients with EOCRC had better survival compared to patients diagnosed at 65 years and above (hazard ratio [HR] 0.73, 95% CI 0.67-0.79, P<.001) after adjusting for covariates. CONCLUSIONS The rise in the incidence of rectal cancer among young adults, especially among Chinese and Malays, in Singapore highlights the need for further research to diagnose CRC earlier and reduce cancer-related morbidity and mortality.
Collapse
Affiliation(s)
- Hui Lionel Raphael Chen
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Qingqing Dawn Chong
- Duke-NUS Medical School, Singapore, Singapore
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Brenda Tay
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Siqin Zhou
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre, Singapore, Singapore
| | - Evelyn Yi Ting Wong
- Duke-NUS Medical School, Singapore, Singapore
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Isaac Seow-En
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Ker Kan Tan
- Division of Colorectal Surgery, Department of Surgery, National University Hospital, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Yi Wang
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Adeline Seow
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Kwong-Wei Emile Tan
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Bee Huat Iain Tan
- Duke-NUS Medical School, Singapore, Singapore
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Sze Huey Tan
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre, Singapore, Singapore
| |
Collapse
|
217
|
Pretzsch E, Peschel CA, Rokavec M, Torlot L, Li P, Hermeking H, Werner J, Klauschen F, Neumann J, Jung A, Kumbrink J. Five-Gene Expression Signature Associated With Acquired FOLFIRI Resistance and Survival in Metastatic Colorectal Cancer. J Transl Med 2025; 105:104107. [PMID: 39954853 DOI: 10.1016/j.labinv.2025.104107] [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: 08/15/2024] [Revised: 01/25/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025] Open
Abstract
FOLFIRI, a combination of folinic acid, 5-fluorouracil, and irinotecan, is one of the recommended first-line chemotherapeutic treatments for metastatic colorectal cancer. Unfortunately, acquired FOLFIRI resistance represents a common obstacle in the treatment of metastatic colorectal cancer patients. Thus, we aimed to identify mechanisms, gene alterations, and gene expression signatures contributing to acquired FOLFIRI resistance by mimicking this problem in a cell culture model and subsequent translation in clinical data sets. Three FOLFIRI-resistant colorectal cancer (CRC) cell lines were established by continuous FOLFIRI treatment. Comparative mutation screening (161 genes) and transcriptomics (pathway and differential expression analyses) were performed in parental and resistant cells. Data reconciliation was performed in GSE62322, a clinical FOLFIRI responder data set (intrinsic resistance). Relapse-free survival (RFS) associations of identified differentially expressed genes and potential gene signatures were investigated in 8 clinical CRC data sets. No mutual genetic alterations were found in FOLFIRI-resistant derivatives. Resistant cell lines displayed activation of mitogen-activated protein kinase, immune response, and epithelial-mesenchymal transition pathways. Twelve differentially expressed genes, significantly differentially expressed in at least 2 of the 3 resistant cell lines, were identified. Comparison with GSE62322 and subsequent survival analyses revealed a 5-gene FOLFIRI signature comprised of CAV2, TNC, TACSTD2, SERPINE2, and PERP that was associated with RFS in multiple data sets including the cancer genome atlas CRC (hazard ratio [HR] =2.634, P = 4.53 × 10-6), in pooled samples of all data sets (all stages [N = 1981]: HR = 1.852, P = 6.44 × 10-13; stage IV [N = 260]: HR = 2.462, P = 5.22 × 10-9). A multivariate Cox regression analysis identified the 5-gene signature as an independent prognostic factor in the cancer genome atlas data set (HR = 1.89, P = .0202). Our analyses revealed a 5-gene FOLFIRI resistance signature associated with RFS that may help predict FOLFIRI resistance and thus avoid unnecessary ineffective treatment. Signature members might also represent targets to fight FOLFIRI resistance.
Collapse
Affiliation(s)
- Elise Pretzsch
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Christiane A Peschel
- Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Matjaz Rokavec
- Experimental and Molecular Pathology, Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Lucien Torlot
- Department of Anaesthesiology, LMU University Hospital, LMU Munich, Germany
| | - Pan Li
- Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Heiko Hermeking
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany; Experimental and Molecular Pathology, Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Frederick Klauschen
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany; Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jens Neumann
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany; Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Andreas Jung
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany; Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jörg Kumbrink
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany; Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
218
|
Yang YM, Kim J, Wang Z, Kim J, Kim SY, Cho GJ, Lee JH, Kim SM, Tsuchiya T, Matsuda M, Pandyarajan V, Pandol SJ, Lewis MS, Gangi A, Noble PW, Jiang D, Merchant A, Posadas EM, Bhowmick NA, Lu SC, You S, Xu AM, Seki E. Metastatic tumor growth in steatotic liver is promoted by HAS2-mediated fibrotic tumor microenvironment. J Clin Invest 2025; 135:e180802. [PMID: 39946200 PMCID: PMC11957696 DOI: 10.1172/jci180802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Steatotic liver enhances liver metastasis of colorectal cancer (CRC), but this process is not fully understood. Steatotic liver induced by a high-fat diet increases cancer-associated fibroblast (CAF) infiltration and collagen and hyaluronic acid (HA) production. We investigated the role of HA synthase 2 (HAS2) in the fibrotic tumor microenvironment in steatotic liver using Has2ΔHSC mice, in which Has2 is deleted from hepatic stellate cells. Has2ΔHSC mice had reduced steatotic liver-associated metastatic tumor growth of MC38 CRC cells, collagen and HA deposition, and CAF and M2 macrophage infiltration. We found that low-molecular weight HA activates Yes-associated protein (YAP) in cancer cells, which then releases connective tissue growth factor to further activate CAFs for HAS2 expression. Single-cell analyses revealed a link between CAF-derived HAS2 and M2 macrophages and CRC cells through CD44; these cells were associated with exhausted CD8+ T cells via programmed death-ligand 1 and programmed cell death protein 1 (PD-1). HA synthesis inhibitors reduced steatotic liver-associated metastasis of CRC, YAP expression, and CAF and M2 macrophage infiltration, and improved response to anti-PD-1 antibody. In conclusion, steatotic liver modulates a fibrotic tumor microenvironment to enhance metastatic cancer activity through a bidirectional regulation between CAFs and metastatic tumors, enhancing the metastatic potential of CRC in the liver.
Collapse
Affiliation(s)
- Yoon Mee Yang
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Pharmacy
- Multidimensional Genomics Research Center, and
- Innovative Drug Development Research Team for Intractable Diseases (BK21 plus), Kangwon National University, Chuncheon, South Korea
| | - Jieun Kim
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Zhijun Wang
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jina Kim
- Samuel Oschin Comprehensive Cancer Institute
- Department of Urology
- Department of Computational Biomedicine, and
| | - So Yeon Kim
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Gyu Jeong Cho
- Department of Pharmacy
- Innovative Drug Development Research Team for Intractable Diseases (BK21 plus), Kangwon National University, Chuncheon, South Korea
| | | | - Sun Myoung Kim
- Department of Pharmacy
- Innovative Drug Development Research Team for Intractable Diseases (BK21 plus), Kangwon National University, Chuncheon, South Korea
| | - Takashi Tsuchiya
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Michitaka Matsuda
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Vijay Pandyarajan
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Stephen J. Pandol
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Michael S. Lewis
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Pathology, Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California, USA
| | | | - Paul W. Noble
- Department of Medicine and Women’s Guild Lung Institute
| | - Dianhua Jiang
- Department of Medicine and Women’s Guild Lung Institute
| | - Akil Merchant
- Samuel Oschin Comprehensive Cancer Institute
- Division of Hematology and Cellular Therapy, Department of Medicine
| | - Edwin M. Posadas
- Samuel Oschin Comprehensive Cancer Institute
- Division of Medical Oncology, Department of Medicine, and
| | - Neil A. Bhowmick
- Samuel Oschin Comprehensive Cancer Institute
- Division of Medical Oncology, Department of Medicine, and
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sungyong You
- Samuel Oschin Comprehensive Cancer Institute
- Department of Urology
- Department of Computational Biomedicine, and
| | - Alexander M. Xu
- Samuel Oschin Comprehensive Cancer Institute
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, Maryland, USA
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Institute
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
219
|
Lim HJ, Park IS, Kim MJ, Seo JW, Ha G, Yang HJ, Jeong DY, Kim SY, Jung CH. Protective Effect of Ganjang, a Traditional Fermented Soy Sauce, on Colitis-Associated Colorectal Cancer in Mice. Foods 2025; 14:632. [PMID: 40002076 PMCID: PMC11854912 DOI: 10.3390/foods14040632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent causes of cancer-related fatalities globally, and its development is closely associated with dietary and microbial factors. The aim of this study was to investigate the potential of ganjang, a traditional fermented soy sauce, in alleviating colitis-associated colorectal cancer (CAC) in a mouse model induced by azoxymethane/dextran sulfate sodium (AOM/DSS). The bacterial composition of ganjang samples from different regions primarily included Lactobacillus spp. and Bacillus spp. Administration of ganjang to AOM/DSS-induced mice significantly improved CAC-related symptoms, including increased body weight, restored colon length, and reduced spleen size. Additionally, ganjang administration led to a decrease in tumor size and number, the modulation of apoptotic and proliferative markers, decreased inflammatory cytokine levels, and the restoration of the intestinal epithelial barrier. Furthermore, ganjang samples altered the gut microbiota composition by increasing the relative abundance of Lactobacillus spp. These findings suggest that ganjang has potential as a functional food for CRC prevention or mitigation, primarily through the modulation of CAC symptoms, tumor growth, inflammatory responses, intestinal epithelial barrier integrity, and gut microbiota composition.
Collapse
Affiliation(s)
- Hyeon-Ji Lim
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Jeonju 54810, Republic of Korea; (H.-J.L.); (I.-S.P.); (M.J.K.); (S.-Y.K.)
| | - In-Sun Park
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Jeonju 54810, Republic of Korea; (H.-J.L.); (I.-S.P.); (M.J.K.); (S.-Y.K.)
| | - Min Ju Kim
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Jeonju 54810, Republic of Korea; (H.-J.L.); (I.-S.P.); (M.J.K.); (S.-Y.K.)
| | - Ji Won Seo
- Microbial Institute for Fermentation Industry, Sunchang 56048, Republic of Korea; (J.W.S.); (G.H.); (H.-J.Y.); (D.-Y.J.)
| | - Gwangsu Ha
- Microbial Institute for Fermentation Industry, Sunchang 56048, Republic of Korea; (J.W.S.); (G.H.); (H.-J.Y.); (D.-Y.J.)
| | - Hee-Jong Yang
- Microbial Institute for Fermentation Industry, Sunchang 56048, Republic of Korea; (J.W.S.); (G.H.); (H.-J.Y.); (D.-Y.J.)
| | - Do-Youn Jeong
- Microbial Institute for Fermentation Industry, Sunchang 56048, Republic of Korea; (J.W.S.); (G.H.); (H.-J.Y.); (D.-Y.J.)
| | - Seon-Young Kim
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Jeonju 54810, Republic of Korea; (H.-J.L.); (I.-S.P.); (M.J.K.); (S.-Y.K.)
| | - Chan-Hun Jung
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Jeonju 54810, Republic of Korea; (H.-J.L.); (I.-S.P.); (M.J.K.); (S.-Y.K.)
| |
Collapse
|
220
|
Bai L, Liu X, Yuan Z, Xu G, Li X, Wan Z, Zhu M, Liang X, Li P, Lan Q, Yu H, Tang G, Huang M, Peng S, Lin J, Wang X, Luo Y, Wei G. Activation of IL-2/IL-2R pathway by Hedyotis diffusa polysaccharide improves immunotherapy in colorectal cancer. Int J Biol Macromol 2025; 306:141013. [PMID: 39954887 DOI: 10.1016/j.ijbiomac.2025.141013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Colorectal cancer (CRC) is a prevalent and highly malignant tumor with a limited response to immune checkpoint inhibitor-based immunotherapy. There is an urgent need for novel immunomodulatory agents to enhance the immunotherapeutic response in CRC. Hedyotis diffusa, known for its immunomodulatory properties, has long been utilized as an adjunct in cancer treatment, positioning it as a potential source for discovering new tumor immunomodulators. In this study, we identified a polysaccharide derived from Hedyotis diffusa (HDP), comprising six monosaccharides: rhamnose, arabinose, galactose, glucose, xylose, and mannose. When combined with PD-1 and CTLA-4 inhibitors, HDP can boost systemic immunity in mice to enhance the effectiveness of immune checkpoint inhibitors in CRC therapy. HDP significantly increases the infiltration of CD4+ and CD8+ T cells into tumor microenvironment and upregulates the expression of key effector molecules derived from cytotoxic T cells. Mechanistic studies reveal that HDP activates the IL-2/IL-2R axis by upregulating IL-2 production and the expression of IL-2 receptor subunits, thereby promoting T cell proliferation. Collectively, this research introduces an innovative strategy to improve the efficacy of tumor immunotherapy by harnessing the immunomodulatory potential of polysaccharides. It also directs a roadmap for developing HDP as a promising immunomodulator for CRC treatment.
Collapse
Affiliation(s)
- Liangliang Bai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Xiaoxia Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Ze Yuan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Gaopo Xu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xuan Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Zhongxian Wan
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei 445000, China
| | - Mingxuan Zhu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xiaoxia Liang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Peisi Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Qiqian Lan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Huichuan Yu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Guannan Tang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Mingzhe Huang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Shaoyong Peng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Jinxing Lin
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xiaolin Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| | - Yanxin Luo
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| | - Gang Wei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
221
|
Ding Q, Li C, Wang C, Ding Q. Construction and interpretation of weight-balanced enhanced machine learning models for predicting liver metastasis risk in colorectal cancer patients. Discov Oncol 2025; 16:164. [DOI: https:/doi.org/10.1007/s12672-025-01871-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/03/2025] [Indexed: 04/06/2025] Open
|
222
|
Avădănei ER, Căruntu ID, Nucă I, Balan RA, Lozneanu L, Giusca SE, Pricope DL, Dascalu CG, Amalinei C. KRAS Mutation Status in Relation to Clinicopathological Characteristics of Romanian Colorectal Cancer Patients. Curr Issues Mol Biol 2025; 47:120. [PMID: 39996841 PMCID: PMC11854687 DOI: 10.3390/cimb47020120] [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: 11/17/2024] [Revised: 02/06/2025] [Accepted: 02/09/2025] [Indexed: 02/26/2025] Open
Abstract
Our study's aim was to evaluate the clinicopathological profile of colorectal cancer (CRC) patients from North-East Romania in relation to the Kirsten rat sarcoma viral oncogene homolog (KRAS). We designed a retrospective study on 108 CRC patients using the fully automated real-time PCR-based molecular testing system, IdyllaTMKRAS Mutation Test (Biocartis, Mechelen, Belgium). Of the patients, 64 (59.3%) were men and 62 (57.4%) were older than the group average, with left bowel location in 38 cases (35.2%), adenocarcinoma NOS in 102 cases (94.4%), mixed histological pattern in 65 cases (60.2%), T3 in 60 patients (55.6%), N2 in 46 patients (42.6%), and 7-12 tumour buds registered in 58 tumours (53.7%). A total of 54 tumour samples (50%) showed KRAS mutation. Statistical comparative analyses associated KRAS mutations with the histopathological pattern (p = 0.018), tumour grade (p = 0.030), depth of invasion (pT) (p < 0.001), lymph node involvement (pN) (p < 0.001), venous vascular invasion (p = 0.048), and tumour buds' number (p = 0.007). Our results demonstrate the relationship between KRAS mutation and clinicopathological features, with possible impact in clinical tumour stratification and therapeutic management.
Collapse
Affiliation(s)
- Elena-Roxana Avădănei
- Department of Morpho-Functional Sciences I-Histology, Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-D.C.); (R.A.B.); (L.L.); (S.-E.G.); (D.L.P.); (C.A.)
- Praxis Medical Investigation Laboratory, 35 Moara de Vant Street, 700376 Iasi, Romania;
| | - Irina-Draga Căruntu
- Department of Morpho-Functional Sciences I-Histology, Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-D.C.); (R.A.B.); (L.L.); (S.-E.G.); (D.L.P.); (C.A.)
- Romanian Medical Science Academy, 1 I.C. Bratianu Boulevard, 030171 Bucharest, Romania
| | - Irina Nucă
- Praxis Medical Investigation Laboratory, 35 Moara de Vant Street, 700376 Iasi, Romania;
- Department of Mother and Child Medicine-Genetics, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Raluca Anca Balan
- Department of Morpho-Functional Sciences I-Histology, Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-D.C.); (R.A.B.); (L.L.); (S.-E.G.); (D.L.P.); (C.A.)
| | - Ludmila Lozneanu
- Department of Morpho-Functional Sciences I-Histology, Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-D.C.); (R.A.B.); (L.L.); (S.-E.G.); (D.L.P.); (C.A.)
- Department of Pathology, “Sf. Spiridon” Clinical Emergency County Hospital, 1 Independentei Street, 700111 Iasi, Romania
| | - Simona-Eliza Giusca
- Department of Morpho-Functional Sciences I-Histology, Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-D.C.); (R.A.B.); (L.L.); (S.-E.G.); (D.L.P.); (C.A.)
| | - Diana Lavinia Pricope
- Department of Morpho-Functional Sciences I-Histology, Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-D.C.); (R.A.B.); (L.L.); (S.-E.G.); (D.L.P.); (C.A.)
| | - Cristina Gena Dascalu
- Department of Preventive Medicine and Interdisciplinarity-Medical Informatics and Biostatistics, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania;
| | - Cornelia Amalinei
- Department of Morpho-Functional Sciences I-Histology, Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-D.C.); (R.A.B.); (L.L.); (S.-E.G.); (D.L.P.); (C.A.)
- Department of Histopathology, Institute of Legal Medicine, 4 Buna Vestire Street, 700455 Iasi, Romania
| |
Collapse
|
223
|
Dumut DC, Hajduch M, Zacharias AM, Duan Q, Frydrych I, Rozankova Z, Popper M, Garic D, Paun RA, Centorame A, Shah J, Mistrik M, Dzubak P, De Sanctis JB, Radzioch D. Diethyldithiocarbamate-copper complex ignites the tumor microenvironment through NKG2D-NKG2DL axis. Front Immunol 2025; 16:1491450. [PMID: 40013140 PMCID: PMC11860975 DOI: 10.3389/fimmu.2025.1491450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/17/2025] [Indexed: 02/28/2025] Open
Abstract
Advanced metastatic colorectal cancer (CRC) with deficient DNA mismatch repair (MMR-d), or immune-hot CRCs, show significantly improved clinical outcomes compared to MMR-proficient (MMR-p), or immune-cold CRCs. While the prior represents about 5% of all CRCs, the latter represent 95% and are characterized by low immunogenicity. This study investigates bis-diethyldithiocarbamate (CuET), a novel anticancer compound, and its impact on the colorectal cancer tumor microenvironment (TME). CuET is shown to convert immunologically inactive tumors into hotbeds of antitumor immune responses, marked by increased lymphocyte infiltration, heightened cytotoxicity of natural killer (NK) and T cells, and enhanced non-self recognition by lymphocytes. The potent anticancer cytotoxicity and in vivo safety and efficacy of CuET are established. In summary, CuET transforms the colorectal cancer TME, bolstering NK and T cell cytotoxicity and refining tumor cell recognition through non-classical activation via the NKG2D/NKG2DL axis. This study unveils a novel mechanism of action for CuET: a potent immunomodulator capable of turning cold tumors hot.
Collapse
Affiliation(s)
- Daciana C. Dumut
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Amanda M. Zacharias
- Department of Biomedical & Molecular Sciences, Faculty of Health Sciences, Queen’s University, Kingston, ON, Canada
| | - Qingling Duan
- Department of Biomedical & Molecular Sciences, Faculty of Health Sciences, Queen’s University, Kingston, ON, Canada
- School of Computing, Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Ivo Frydrych
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
| | - Zuzana Rozankova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Miroslav Popper
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Dusan Garic
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Radu Alexandru Paun
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Amanda Centorame
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
| | - Juhi Shah
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
| | - Martin Mistrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Petr Dzubak
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Juan B. De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Danuta Radzioch
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| |
Collapse
|
224
|
Ding Q, Li C, Wang C, Ding Q. Construction and interpretation of weight-balanced enhanced machine learning models for predicting liver metastasis risk in colorectal cancer patients. Discov Oncol 2025; 16:164. [PMID: 39937330 PMCID: PMC11822177 DOI: 10.1007/s12672-025-01871-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a major contributor to cancer-related mortality, with liver metastases developing in approximately 25% of affected individuals. The presence of liver metastasis significantly deteriorates the prognosis for patients. The objective of this study is to predict liver metastasis in CRC patients by developing machine learning (ML)-based models, thereby aiding clinicians in the decision-making process for appropriate interventions. METHODS Retrospective analysis was performed using the Surveillance, Epidemiology, and End Results (SEER) database, and cases with CRC from 2010 to 2015 were extracted to the downstream analysis. Logistic regression (LR), Random Forest (RF), Gradient Boosting Machine (GBM), Extreme Gradient Boosting (XGBoost), Categorical Boosting (CatBoost), and LightGBM are applied to develop machine learning (ML) models to predict liver metastasis of CRC patient. To optimize the models, an improved weight-balancing algorithm was employed, enhancing the performance of the classifiers. The six models were tenfold cross-validated, and the optimal model was selected based on a combination of performance metrics. Shapley additive explanation (SHAP) was utilized to interpret the best-performing ML models globally, locally, and interactively. To ensure the model's reliability and generalizability, an external validation cohort of CRC cases from 2018 to 2021, obtained from a separate SEER database, was used for external evaluation. RESULTS In total, 50,062 patients with CRC were included in the analysis, with 5604 patients occurring liver metastasis. Among the six models evaluated, the CatBoost model showed excellent performance with the highest AUC of 0.8844. Moreover, the CatBoost model also outperformed the others in terms of recall (0.8060) and F1-score (0.6736). SHAP-based summary and force plots were used to interpret the CatBoost model. The interpretability analysis revealed that elevated carcinoembryonic antigen (CEA) levels, systemic therapy, N and T stages, and chemotherapy performed were the most significant indicators for predicting liver metastasis according to the optimal model. Furthermore, systemic therapy was suggested to increase liver metastasis risk in N0 stage patients, while it appeared to be beneficial in patients with lymph node metastasis. Preoperative radiation therapy was found to be more effective than postoperative radiation therapy. Validation using an external cohort of CRC cases from 2018 to 2021 further confirmed the robustness and stability of the CatBoost model, as its overall performance remained consistent with the internal validation results. CONCLUSION Elevated levels of carcinoembryonic antigen (CEA) have been identified as a crucial clinical predictor for liver metastasis in CRC patients. Furthermore, the administration of systemic therapy to patients who do not exhibit lymph node involvement has been found to increase the risk of liver metastasis. In terms of radiation therapy, preoperative radiation appears to be more efficacious in controlling the risk of liver metastasis compared to postoperative radiation. This finding underscores the importance of optimizing treatment strategies based on the specific clinical context and patient characteristics.
Collapse
Affiliation(s)
- Qunzhe Ding
- School of Information Management, Wuhan University, Wuhan, Hubei, 430072, People's Republic of China
| | - Chenyang Li
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chendong Wang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qunzhe Ding
- School of Information Management, Wuhan University, Wuhan, Hubei, 430072, People's Republic of China.
| |
Collapse
|
225
|
Wang Y, Qiu X, Li Q, Qin J, Ye L, Zhang X, Huang X, Wen X, Wang Z, He W, Di Y, Zhou Q. Single-cell and spatial-resolved profiling reveals cancer-associated fibroblast heterogeneity in colorectal cancer metabolic subtypes. J Transl Med 2025; 23:175. [PMID: 39934919 DOI: 10.1186/s12967-025-06103-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 01/08/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) presents significant treatment challenges due to its high heterogeneity and complex intercellular interactions. Further exploration of CRC subtypes and interactions among tumor-specific clusters will facilitate the development of personalized treatment strategies. METHODS Single-cell RNA sequencing and bulk RNA sequencing datasets were integrated to determine CRC metabolic subtypes by hierarchical clustering. The analysis was further extended to cellchat, pseudotime, immune infiltration, and clinicopathological relevance to explore the characteristics of secreted frizzled related protein 2 (SFRP2) + cancer-associated fibroblast (CAF) clusters, and validated by spatial transcriptomics (ST), in vivo experiments, and multiple immunohistochemistry (mIHC). RESULTS CRC samples were stably classified into three heterogeneous metabolic subtypes, each exhibiting different microenvironment and CAF heterogeneity, particularly in the distribution of SFRP2 + CAF, which was aligned with metabolic activity. SFRP2 + CAF exhibits high extracellular matrix (ECM) activity and is closely involved in cellular communication, not only promoting the malignant progression of cancer cells but also inducing the differentiation of Tregs. Compared to responders of chemotherapy, the proportion of SFRP2 + CAFs is significantly increased in non-responders. Importantly, mIHC and ST analyses confirm that cancer cells with low expression of agmatinase (AGMAT) can recruit SFRP2 + CAFs, and Treg infiltration surrounding SFRP2 + CAFs was observed. AGMAT combined with oxaliplatin showed the best efficacy in vivo, which may be associated with the inhibition of SFRP2 + CAF infiltration. CONCLUSIONS Our study identified and described the potential protumor biological properties of SFRP2 + CAFs, and AGMAT may be a valuable target for disrupting their properties.
Collapse
Affiliation(s)
- Youpeng Wang
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Xingfeng Qiu
- Department of Gastrointestinal Surgery, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361000, China
| | - Qinghai Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Jiale Qin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Lvlan Ye
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xiang Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Xingxiang Huang
- Department of Gastrointestinal Surgery, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361000, China
| | - Xiangqiong Wen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Ziyang Wang
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Weiling He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China.
- Department of Gastrointestinal Surgery, School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361000, China.
| | - Yuqin Di
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China.
- Molecular Diagnosis and Gene Testing Center, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China.
| | - Qi Zhou
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China.
- Department of General Surgery, Hui Ya Hospital of The First Affiliated Hospital, Sun Yat-sen University, Huizhou, Guangdong, 516081, China.
| |
Collapse
|
226
|
Zhang H, Tang J, Cao H, Wang C, Shen C, Liu J. Effect and mechanism of Magnolia officinalis in colorectal cancer: Multi-component-multi-target approach. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119007. [PMID: 39471878 DOI: 10.1016/j.jep.2024.119007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/13/2024] [Accepted: 10/26/2024] [Indexed: 11/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Colorectal cancer (CRC) is a prevalent malignant tumor of the digestive tract. Traditional Chinese medicine (TCM) has a long history of treating CRC, with advantages such as effectiveness, multi-target, multi-pathway, and minimal side effects. TCM Magnolia officinalis (M. officinalis) refers to the dried bark, root bark, and branch bark of either Magnolia officinalis Rehd.et Wils. or Magnolia officinalis Rehd.et Wils. var. biloba Rehd.et Wils. It is commonly utilized to alleviate the side effects of chemotherapy for CRC, owing to its anti-inflammatory and anti-tumor properties. However, current research primarily focuses on the individual components and does not take into consideration the characteristics of multi-component-multi-target action. AIM OF THE STUDY Our aim is to study the new action characteristics of M. officinalis in the treatment of CRC. MATERIALS AND METHODS Utilizing network pharmacology to identify potential active ingredients, key targets, and main signaling pathways of M. officinalis for the treatment of CRC. The binding effect was further validated through molecular docking analysis. Furthermore, the aforementioned components were identified using liquid chromatography-mass spectrometry (LC-MS), and the cleavage pathways of the main components were analyzed. Subsequently, both in vitro and in vivo experiments were carried out to investigate the anti-CRC effect of the active ingredients of M. officinalis and its potential mechanism. RESULTS Network pharmacology and Molecular docking identified 5 main active ingredients and 6 core targets of M. officinalis for the treatment of CRC. Then, LC-MS identified the active components of M. officinalis. At the same time, both in vitro and in vivo experiments have confirmed the ability of Eucalyptol (Euc) and Obovatol (Obo)to inhibit inflammation and tumor cell proliferation. The possible mechanism involved is that Euc and Obo counteract CRC by inhibiting the over-activation of NF-κBp65/JAK and Bcl-2/Caspase signaling pathways, respectively. They also play a role in the anti-CRC effect of M. officinalis. CONCLUSION Magnolol (MAG), Honokiol (HK), Euc, Obo, and Neohesperidin (NHP) in M. officinalis may be the pharmacological substance basis for its anti-cancer effect on CRC. The treatment of CRC with M. officinalis is characterized by its multi-component, multi-target, and multi-pathway approach. These findings provide a theoretical basis for further inspiring the clinical application of M. officinalis and the development of efficacy targets.
Collapse
Affiliation(s)
- He Zhang
- State Key Laboratory of Dynamic Measurement Technology, School of Instrument and Electronics, North University of China, Taiyuan, 030051, China
| | - Jun Tang
- State Key Laboratory of Dynamic Measurement Technology, School of Instrument and Electronics, North University of China, Taiyuan, 030051, China
| | - Huiliang Cao
- State Key Laboratory of Dynamic Measurement Technology, School of Instrument and Electronics, North University of China, Taiyuan, 030051, China
| | - Chenguang Wang
- State Key Laboratory of Dynamic Measurement Technology, School of Instrument and Electronics, North University of China, Taiyuan, 030051, China
| | - Chong Shen
- State Key Laboratory of Dynamic Measurement Technology, School of Instrument and Electronics, North University of China, Taiyuan, 030051, China.
| | - Jun Liu
- State Key Laboratory of Dynamic Measurement Technology, School of Instrument and Electronics, North University of China, Taiyuan, 030051, China.
| |
Collapse
|
227
|
Wang C, Huang T, Wang X. Efficacy and safety of transanal endoscopic microsurgery for early rectal cancer: a meta-analysis. Front Oncol 2025; 15:1545547. [PMID: 39995839 PMCID: PMC11847824 DOI: 10.3389/fonc.2025.1545547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 01/17/2025] [Indexed: 02/26/2025] Open
Abstract
Background The gold standard for the treatment of rectal cancer is radical surgery with total mesorectal excision (TME). As one of the alternatives to radical surgery, local resection has been proposed for the treatment of early rectal cancer. The purpose of this article was to evaluate the safety and efficacy of transanal endoscopic microsurgery (TEM) in the treatment of early rectal cancer. Methods By searching the PubMed, Cochrane Library, Web of Science, and China National Knowledge Infrastructure databases, we selected all articles on TEM for early rectal cancer. Two researchers independently completed the entire process from screening, inclusion to data extraction and performed statistical analysis using RevMan 5.3. The primary outcomes included basic patient characteristics, overall survival rate, disease-free survival rate, disease-specific survival rate, recurrence rate, and complication rate and type. Results A total of 33 articles were included in this meta-analysis. The results showed that the overall survival rate was 100% for T0 stage, 98.1% for Tis (carcinoma in situ) stage, and 80.2% for early stage rectal cancer patients (83.9% for T1 and 72.4% for T2). The weighted overall survival rate was 94% (RD = 0.94, 95% CI = 0.93-0.95, I 2 = 80%, P < 0.00001) for all stage patients, the weighted disease-free survival rate was 91% (RD = 0.91, 95% CI = 0.90-0.93, I 2 = 83%, P < 0.00001), and the disease-specific survival rate was 97% (RD = 0.97, 95% CI = 0.96-0.98, I 2 = 63%, P < 0.00001). The recurrence rate was 0.5% for T0 stage, 1.9% for Tis stage, and 11.9% for early stage rectal cancer patients (8.1% for T1 and 19.7% for T2). The weighted recurrence rate was 7% (RD = 0.07, 95% CI = 0.06-0.08, I 2 = 69%, P < 0.00001) for all stage patients. The weighted complications rate was 11% (RD = 0.11, 95% CI = 0.10-0.12, I 2 = 66%, P < 0.00001) for all stage patients, with Clavien-Dindo grade I accounting for 77.7%, Clavien-Dindo grade II accounting for 8%, and Clavien-Dindo grade III accounting for 14.3%. Conclusion The results showed that TEM has a high postoperative survival rate, low recurrence rate, and low complication rate in the T0 stage, Tis stage, and T1 stage, indicating its good safety and efficacy. For the treatment of T2 stage, TEM has a lower overall survival rate and a higher recurrence rate. Our meta-analysis results suggest that TEM alone is not recommended as a curative treatment for T2 stage; on the contrary, TME is more frequently recommended.
Collapse
Affiliation(s)
- Chunqiang Wang
- Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Tianye Huang
- The First Medical University of Shandong Affiliated Linyi Hospital, Linyi, Shandong, China
| | - Xuebing Wang
- The First Medical University of Shandong Affiliated Taian Hospital, Taian, Shandong, China
| |
Collapse
|
228
|
Yeo MR, Voutsadakis IA. Characteristics, Therapeutic Approaches and Outcomes of Patients Older than 80 Years Old with Metastatic Colorectal Cancer Compared with Younger Patients. J Clin Med 2025; 14:1099. [PMID: 40004631 PMCID: PMC11856852 DOI: 10.3390/jcm14041099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/31/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background: While advances in therapies have improved metastatic cancer survival rates, elderly patients with colorectal cancer often experience delayed diagnoses, receive less frequent systemic therapies, and show inferior survival outcomes compared to younger groups. Patients over the age of 80 years old face greater treatment risks due to frailty and comorbidities. In this article, we examine characteristics, treatment and outcomes in older adults with metastatic colorectal cancer. Methods: The medical records of all patients aged 80 years and above and comparable patients aged 65-75 years old, who were diagnosed with stage 4 colorectal cancer at a cancer center over a six-year period, were retrospectively reviewed. Results: Patients in the 80 years old and older group more frequently had right-sided primary colon cancer (71.5%), compared to younger patients aged 65-75 years old (34.1%, p = 0.006). Patients in the younger cohort more commonly presented with stage 4 disease at initial diagnosis (59.5%) compared to older patients (22.2%). Elevated carcinoembryonic antigen (CEA) levels were more commonly identified in younger metastatic patients (76.3% vs. 46.4%, p = 0.013). Patients in the younger age group were more likely to have received previous neoadjuvant and adjuvant chemotherapy prior to metastatic progression (p = 0.02, and p = 0.01); however, a significant difference in palliative chemotherapy was not identified between the age groups of metastatic patients. The adverse effects of chemotherapy treatment were similar between the age groups. Conclusions: The active treatment of metastatic colorectal cancer in patients aged 80 and above is feasible when tailored according to the patients' performance status, comorbidities, and life expectancy. Understanding metastatic disease presentations in elderly patients can improve treatment outcomes in this challenging-to-treat group.
Collapse
Affiliation(s)
- Melissa R. Yeo
- Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada;
| | - Ioannis A. Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste. Marie, ON P6B 0A8, Canada
- Section of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
229
|
Zhang F, Chen X, Qiao C, Yang S, Zhai Y, Zhang J, Chai K, Wang H, Zhou J, Guo M, Lu P, Wu J. Exploring the Anti-Colorectal Cancer Mechanism of Norcantharidin Through TRAF5/NF-κB Pathway Regulation and Folate-Targeted Liposomal Delivery. Int J Mol Sci 2025; 26:1450. [PMID: 40003916 PMCID: PMC11855010 DOI: 10.3390/ijms26041450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 01/26/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Colorectal cancer is one of the most common malignant tumors worldwide, significantly impacting human health. Cantharidin (CTD), an active compound derived from the Spanish fly, exhibits antitumor properties. Its derivative, norcantharidin (NCTD), is synthesized by removing methyl groups from positions 1 and 2 of cantharidin. NCTD has demonstrated lower toxicity while maintaining similar antitumor effects compared to CTD. However, the mechanism by which NCTD exerts its effects against colorectal cancer remains unclear. Here, we conducted a comprehensive analysis of the effects of NCTD on colorectal cancer both in vitro and in vivo. Whole-transcriptome sequencing and bioinformatics tools were employed to identify potential key targets of NCTD in the treatment of colorectal cancer. Additionally, we designed folate-receptor-targeting NCTD liposomes (FA-NCTD) and assessed their anticancer efficacy in vivo. NCTD effectively inhibited cell viability, clonal formation, and migration in HCT116 and HT-29 cell lines. NCTD also induced apoptosis, influenced the cell cycle, altered mitochondrial membrane potential, and increased reactive oxygen species levels. The whole-transcriptome sequencing and bioinformatics analysis identified TRAF5 as a key target for NCTD's action against colorectal cancer. Furthermore, NCTD was found to regulate the TRAF5/NF-κB signaling pathway in both HCT116 and HT-29 cells. The FA-NCTD liposomes demonstrated effective tumor targeting and significantly inhibited tumor growth in vivo. This result showed that NCTD effectively suppresses the malignant proliferation of colon cancer cells by modulating the TRAF5/NF-κB signaling pathway and inducing programmed apoptosis, thereby offering a novel strategy for colorectal cancer treatment. The prepared FA-NCTD liposomes provide a promising approach for achieving the precise targeting and controlled release of NCTD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (F.Z.); (X.C.); (C.Q.); (S.Y.); (Y.Z.); (J.Z.); (K.C.); (H.W.); (J.Z.); (M.G.); (P.L.)
| |
Collapse
|
230
|
Gustav M, van Treeck M, Reitsam NG, Carrero ZI, Loeffler CML, Meneghetti AR, Märkl B, Boardman LA, French AJ, Goode EL, Gsur A, Brezina S, Gunter MJ, Murphy N, Hönscheid P, Sperling C, Foersch S, Steinfelder R, Harrison T, Peters U, Phipps A, Kather JN. Assessing Genotype-Phenotype Correlations with Deep Learning in Colorectal Cancer: A Multi-Centric Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.04.25321660. [PMID: 39973981 PMCID: PMC11838662 DOI: 10.1101/2025.02.04.25321660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Background Deep Learning (DL) has emerged as a powerful tool to predict genetic biomarkers directly from digitized Hematoxylin and Eosin (H&E) slides in colorectal cancer (CRC). However, few studies have systematically investigated the predictability of biomarkers beyond routinely available alterations such as microsatellite instability (MSI), and BRAF and KRAS mutations. Methods Our primary dataset comprised H&E slides of CRC tumors across five cohorts totaling 1,376 patients who underwent comprehensive panel sequencing, with an additional 536 patients from two public datasets for validation. We developed a DL model using a single transformer model to predict multiple genetic alterations directly from the slides. The model's performance was compared against conventional single-target models, and potential confounders were analyzed. Findings The multi-target model was able to predict numerous biomarkers from pathology slides, matching and partly exceeding single-target transformers. The Area Under the Receiver Operating Characteristic curve (AUROC, mean ± std) on the primary external validation cohorts was: BRAF (0·78 ± 0·01), hypermutation (0·88 ± 0·01), MSI (0·93 ± 0·01), RNF43 (0·86 ± 0·01); this biomarker predictability was mirrored across metrics and co-occurrence analyses. However, biomarkers with high AUROCs largely correlated with MSI, with model predictions depending considerably on MSI-associated morphology upon pathological examination. Interpretation Our study demonstrates that multi-target transformers can predict the biomarker status for numerous genetic alterations in CRC directly from H&E slides. However, their predictability is mainly associated with MSI phenotype, despite indications of slight biomarker-inherent contributions to a phenotype. Our findings underscore the need to analyze confounders in AI-based oncology biomarkers. To enable this, we developed a validated model applicable to other cancers and larger, diverse datasets. Funding The German Federal Ministry of Health, the Max-Eder-Programme of German Cancer Aid, the German Federal Ministry of Education and Research, the German Academic Exchange Service, and the EU.
Collapse
Affiliation(s)
- Marco Gustav
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
| | - Marko van Treeck
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
| | - Nic G. Reitsam
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Zunamys I. Carrero
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
| | - Chiara M. L. Loeffler
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
- Department of Medicine I, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
| | - Asier Rabasco Meneghetti
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
| | - Bruno Märkl
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Lisa A. Boardman
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Amy J. French
- Division of Laboratory Genetics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ellen L. Goode
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea Gsur
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Stefanie Brezina
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Marc J. Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
- Cancer Epidemiology and Prevention Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Neil Murphy
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Pia Hönscheid
- Institute of Pathology, University Hospital Carl Gustav Carus (UKD), Technical University Dresden (TUD), Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, German Cancer Research Center Heidelberg, Dresden, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Sperling
- Institute of Pathology, University Hospital Carl Gustav Carus (UKD), Technical University Dresden (TUD), Dresden, Germany
| | - Sebastian Foersch
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Robert Steinfelder
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tabitha Harrison
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Amanda Phipps
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
- Department of Medicine I, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James’s, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
231
|
Smeu A, Marcovici I, Dehelean CA, Dumitrel SI, Borza C, Lighezan R. Flavonoids and Flavonoid-Based Nanopharmaceuticals as Promising Therapeutic Strategies for Colorectal Cancer-An Updated Literature Review. Pharmaceuticals (Basel) 2025; 18:231. [PMID: 40006045 PMCID: PMC11858883 DOI: 10.3390/ph18020231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Colorectal cancer (CRC) represents one of the most serious health issues and the third most commonly diagnosed cancer worldwide. However, the treatment options for CRC are associated with adverse reactions, and in some cases, resistance can develop. Flavonoids have emerged as promising alternatives for CRC prevention and therapy due to their multitude of biological properties and ability to target distinct processes involved in CRC pathogenesis. Their innate disadvantageous properties (e.g., low solubility and stability, reduced bioavailability, and lack of tumor specificity) have delayed the potential inclusion of flavonoids in CRC treatment regimens but have hastened the design of nanopharmaceuticals comprising a flavonoid agent entrapped in a nanosized delivery platform that not only counteract these inconveniences but also provide an augmented therapeutic effect and an elevated safety profile by conferring a targeted action. Starting with a brief presentation of the pathological features of CRC and an overview of flavonoid classes, the present study comprehensively reviews the anti-CRC activity of different flavonoids from a mechanistic perspective while also portraying the latest discoveries made in the area of flavonoid-containing nanocarriers that have proved efficient in CRC management. This review concludes by showcasing future perspectives for the advancement of flavonoids and flavonoid-based nanopharmaceuticals in CRC research.
Collapse
Affiliation(s)
- Andreea Smeu
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Iasmina Marcovici
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Cristina Adriana Dehelean
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Stefania-Irina Dumitrel
- Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Claudia Borza
- Department of Functional Sciences, Discipline of Pathophysiology, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timișoara, Romania
- Centre for Translational Research and Systems Medicine, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timișoara, Romania
- Centre of Cognitive Research in Pathological Neuro-Psychiatry NEUROPSY-COG, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Rodica Lighezan
- Center for Diagnosis and Study of Parasitic Diseases, Department of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Discipline of Parasitology, Department of Infectious Diseases, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
232
|
Lyu H, Chen X, Cheng Y, Zhang T, Wang P, Wong JHY, Wang J, Stasiak L, Sun L, Yang G, Wang L, Yue F. Pioneer factor GATA6 promotes colorectal cancer through 3D genome regulation. SCIENCE ADVANCES 2025; 11:eads4985. [PMID: 39919174 PMCID: PMC11804904 DOI: 10.1126/sciadv.ads4985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/09/2025] [Indexed: 02/09/2025]
Abstract
Colorectal cancer (CRC) is one of the most lethal and prevalent malignancies. While the overexpression of pioneer factor GATA6 in CRC has been linked with metastasis, its role in genome-wide gene expression dysregulation remains unclear. Through studies of primary human CRC tissues and analysis of the TCGA data, we found that GATA6 preferentially binds at CRC-specific active enhancers, with enrichment at enhancer-promoter loop anchors. GATA6 protein also physically interacts with CTCF, suggesting its critical role in 3D genome organization. The ablation of GATA6 through AID and CRISPR systems severely impaired cancer cell clonogenicity and proliferation. Mechanistically, GATA6 knockout induced global loss of CRC-specific open chromatins and extensive alterations of critical enhancer-promoter interactions for CRC oncogenes. Last, we showed that GATA6 knockout greatly reduced tumor growth and improved survival in mice. Together, we revealed a previously unidentified mechanism by which GATA6 contributes to the pathogenesis of colorectal cancer.
Collapse
Affiliation(s)
- Huijue Lyu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Xintong Chen
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yang Cheng
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Te Zhang
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ping Wang
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Josiah Hiu-yuen Wong
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Juan Wang
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lena Stasiak
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Leyu Sun
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Guangyu Yang
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Lu Wang
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Feng Yue
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|
233
|
Abrantes AM, Caetano-Oliveira R, Oliveiros B, Cipriano MA, Tralhão JG. Association Between Colorectal Cancer Primary Features and Liver Metastases Histological Growth Patterns: Inflammation on the Primary Tumor is Associated with Desmoplastic Growth Pattern. Clin Colorectal Cancer 2025:S1533-0028(25)00016-7. [PMID: 40021416 DOI: 10.1016/j.clcc.2025.01.004] [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: 06/03/2024] [Revised: 01/04/2025] [Accepted: 01/29/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND More than 50% of patients diagnosed with colorectal cancer (CRC) will develop liver metastases (CRCLM), which is the main cause of death for more than 60% of these patients. The aim of this study was to correlate the clinical and pathological characteristics of the primary CRC and CRCLM, with emphasis in predicting the histological growth pattern of the CRCLM. METHODS Cohort of 73 patients with CRC. Analysis of clinical data and blinded pathological review was performed related with primary tumor and CRCLM features. The analysis was performed in SPSS (version 27) with a significance level of 5%. RESULTS A statistically significant association was found between tumor size and metastasis growth pattern (P = .002), with larger tumors giving rise to metastases with a nondesmoplastic growth pattern. Lymphovascular invasion (LVI) was associated with metachronous CRCLM (P = .043). In the absence of LVI, the time required for CRCLM to appear was significantly longer (P = .011). The number of metastases was significantly higher (P = .049) in tumors without LVI when compared to tumors with LVI. There was a statistically significant association between CRC high-grade inflammation and the desmoplastic metastases growth pattern of the CRCLM (P = .017). CONCLUSION The possibility of predicting the CRCLM histological growth pattern resorting to primary CRC characteristics would be useful for proper patient selection for surgery and adapting biological therapies.
Collapse
Affiliation(s)
- Ana Margarida Abrantes
- Biophysics Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal; Coimbra Centor Académico e Clínico (CACC), Coimbra, Portugal
| | - Rui Caetano-Oliveira
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal; Coimbra Centor Académico e Clínico (CACC), Coimbra, Portugal; Pathology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal; Centro de Anatomia Patológica Germano de Sousa, Coimbra, Portugal; General Surgery Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.
| | - Bárbara Oliveiros
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal; Coimbra Centor Académico e Clínico (CACC), Coimbra, Portugal
| | | | - José Guilherme Tralhão
- Biophysics Institute, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal; Coimbra Centor Académico e Clínico (CACC), Coimbra, Portugal; General Surgery Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| |
Collapse
|
234
|
Hao C, Pu Y, Li J, Zhong Z, Huang Z, Wang X. CircWDR78 inhibits the development of colorectal cancer by regulating the miR-653-3p/RGS4 axis. J Cancer Res Clin Oncol 2025; 151:66. [PMID: 39912944 PMCID: PMC11802675 DOI: 10.1007/s00432-025-06092-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/12/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) stands as one of the most serious threats to human health, with its mortality rate increase during the past years. Low diagnostic accuracy, poor prognosis, and high recurrence rates attributed to the high mortality rate of CRC. Consequently, the urgency to identify potential diagnose and biological targets for intervention in this disease. Among the various molecular factor associated with this disease, circular RNAs (circRNAs) have been a hot topic. These noncoding RNA molecules, characterized by their unique closed loop structure, displayed close associations with the progression of tumors. METHODS Actinomycin D experiment and RNA digestion experiment were used to verify the circular structure characteristics of circWDR78. Proliferation and motility ability of circWDR78 was evaluated by in vitro functional experiment. We also used RNA-seq technology to explore the signal pathways and genes it might regulate. Finally, the luciferase assay and qRT-PCR experiment proved that circWDR78 could sponge miR-653-3p, and confirmed that RGS4 is the downstream target of miR-653-3p. RESULTS This study demonstrated that circWDR78 is lower expression in colorectal cancer tissues, revealing its capacity to inhibit proliferation, colony forming ability and cell motility. These findings hint at a potential correlation between its downregulation and the progression of CRC. Mechanistically, we found that circWDR78 could sponge miR-653-3p and identified RGS4 as a novel target of miR-653-3p. CONCLUSION This discovery highlights the significance of circWDR78 as a potential regulatory axis of miR-653-3p/RGS4 in CRC progression.
Collapse
Affiliation(s)
- Chu Hao
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yunju Pu
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Jiunian Li
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Zhi Zhong
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Zhaohui Huang
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
| | - Xue Wang
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China.
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China.
| |
Collapse
|
235
|
Wang S, Wu X, Bi W, Xu J, Hou L, Li G, Pan Y, Zhang H, Li M, Du S, Zhang M, Liu D, Jin S, Shi X, Tian Y, Shuai J, Plikus MV, Song M, Zhou Z, Yu L, Lv C, Yu Z. ROS-induced cytosolic release of mitochondrial PGAM5 promotes colorectal cancer progression by interacting with MST3. Nat Commun 2025; 16:1406. [PMID: 39915446 PMCID: PMC11802746 DOI: 10.1038/s41467-025-56444-2] [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: 02/24/2024] [Accepted: 01/18/2025] [Indexed: 02/09/2025] Open
Abstract
Aberrant release of mitochondrial reactive oxygen species (mtROS) in response to cellular stress is well known for promoting cancer progression. However, precise molecular mechanism by which mtROS contribute to epithelial cancer progression remains only partially understood. Here, using colorectal cancer (CRC) models, we show that upon sensing excessive mtROS, phosphatase PGAM5, which normally localizes to the mitochondria, undergoes aberrant cleavage by presenilin-associated rhomboid-like protein (PARL), becoming released into the cytoplasm. Cytosolic PGAM5 then directly binds to and dephosphorylates MST3 kinase. This, in turn, prevents STK25-mediated LATS1/2 phosphorylation, leading to YAP activation and CRC progression. Importantly, depletion of MST3 reciprocally promotes accumulation of cytosolic PGAM5 by inducing mitochondrial damage. Taken together, these findings demonstrate how mtROS promotes CRC progression by activating YAP via a post-transcriptional positive feedback loop between PGAM5 and MST3, both of which can serve as potential targets for developing next-generation anti-colon cancer therapeutics.
Collapse
Affiliation(s)
- Shiyang Wang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xi Wu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Wenxin Bi
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jiuzhi Xu
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Liyuan Hou
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Guilin Li
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yuwei Pan
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Hanfu Zhang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Mengzhen Li
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Sujuan Du
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Mingxin Zhang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Di Liu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Shuiling Jin
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojing Shi
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuhua Tian
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jianwei Shuai
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Zhaocai Zhou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lu Yu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China.
| | - Cong Lv
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| | - Zhengquan Yu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
236
|
Li Y, Li S, Xiao R, Li X, Yi Y, Zhang L, Zhou Y, Wan Y, Wei C, Zhong L, Yang W, Yao L. A pelvis MR transformer-based deep learning model for predicting lung metastases risk in patients with rectal cancer. Front Oncol 2025; 15:1496820. [PMID: 39980546 PMCID: PMC11841465 DOI: 10.3389/fonc.2025.1496820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Objective Accurate preoperative evaluation of rectal cancer lung metastases (RCLM) is critical for implementing precise medicine. While artificial intelligence (AI) methods have been successful in detecting liver and lymph node metastases using magnetic resonance (MR) images, research on lung metastases is still limited. Utilizing MR images to classify RCLM could potentially reduce ionizing radiation exposure and the costs associated with chest CT in patients without metastases. This study aims to develop and validate a transformer-based deep learning (DL) model based on pelvic MR images, integrated with clinical features, to predict RCLM. Methods A total of 819 patients with histologically confirmed rectal cancer who underwent preoperative pelvis MRI and carcinoembryonic antigen (CEA) tests were enrolled. Six state-of-the-art DL methods (Resnet18, EfficientNetb0, MobileNet, ShuffleNet, DenseNet, and our transformer-based model) were trained and tested on T2WI and DWI to predict RCLM. The predictive performance was assessed using the receiver operating characteristic (ROC) curve. Results Our transformer-based DL model achieved impressive results in the independent test set, with an AUC of 83.74% (95% CI, 72.60%-92.83%), a sensitivity of 80.00%, a specificity of 78.79%, and an accuracy of 79.01%. Specifically, for stage T4 and N2 rectal cancer cases, the model achieved AUCs of 96.67% (95% CI, 87.14%-100%, 93.33% sensitivity, 89.04% specificity, 94.74% accuracy), and 96.83% (95% CI, 88.67%-100%, 100% sensitivity, 83.33% specificity, 88.00% accuracy) respectively, in predicting RCLM. Our DL model showed a better predictive performance than other state-of-the-art DL methods. Conclusion The superior performance demonstrates the potential of our work for predicting RCLM, suggesting its potential assistance in personalized treatment and follow-up plans.
Collapse
Affiliation(s)
- Yin Li
- Department of Information, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Information, The Sixth Affiliated Hospital, Sun Yat-sen University Yuexi Hospital, Maoming, China
| | - Shuang Li
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of General Practice, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruolin Xiao
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Guangzhou, China
| | - Xi Li
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University Yuexi Hospital, Maoming, China
| | - Yongju Yi
- Department of Information, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liangyou Zhang
- Department of Information, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - You Zhou
- Department of Information, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yun Wan
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University Yuexi Hospital, Maoming, China
| | - Chenhua Wei
- Department of Information, The Sixth Affiliated Hospital, Sun Yat-sen University Yuexi Hospital, Maoming, China
| | - Liming Zhong
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Guangzhou, China
| | - Wei Yang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Guangzhou, China
| | - Lin Yao
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Information, The Sixth Affiliated Hospital, Sun Yat-sen University Yuexi Hospital, Maoming, China
- Department of General Practice, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
237
|
Pappas L, Quintanilha JCF, Huang RSP, Parikh AR. Genomic alterations associated with early-stage disease and early recurrence in patients with colorectal cancer. Oncologist 2025; 30:oyae269. [PMID: 39531357 PMCID: PMC11883158 DOI: 10.1093/oncolo/oyae269] [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: 04/18/2024] [Accepted: 08/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The molecular characterization of early-stage (1-3) colorectal cancer (CRC) remains incomplete, as opposed to metastatic disease, where comprehensive genomic profiling (CGP) is routinely performed. This study aimed to characterize the genomics of stages 1-3 versus IV CRC, and the genomics of patients recurring within 1 year of diagnosis. PATIENTS AND METHODS Patients from a de-identified CRC clinico-genomic database who received Foundation Medicine testing (FoundationOne/FoundationOne CDx) during routine clinical care at approximately 280 US cancer clinics between March 2014 and June 2023 were included. Genomic alterations (GA) were compared by Fisher's exact test. RESULTS A total of 4702 patients were included; 1902 with stages 1-3 and 2800 with stage 4 disease. Among patients with stages 1-3 disease, 546 recurred within 1 year. Patients staged 1-3 had higher prevalence of microsatellite instability (MSI-H, 11.4% vs 4.5%, P < .001), tumor mutational burden (TMB) ≥ 10 Mut/Mb (14.6% vs 6.8%, P < .001), GA in RNF43 (11.2% vs 5.7%, P < .001), MSH6 (3.9% vs 1.7%, P < .001), MLH1 (2.3% vs 0.7%, P < .001), and MSH2 (1.5% vs 0.6%, P < .01) compared to those with stage 4 disease. Patients who recurred within 1 year had higher prevalence of MSI-H (13.2% vs 4.4%, P < .001), TMB ≥ 10 Mut/Mb (16.2% vs 6.9%, P < .001), BRAF V600E (17.2% vs 7.9%, P < .003), GA in RNF43 (13.7% vs 5.3%, P < .001), MSH6 (4.2% vs 1.6%, P = .035), and BRCA1/2 (6.2% vs 3.0%, P = .030). On recurrence, more patients received targeted therapy when CGP was performed before versus after first-line therapy (43% vs 19%, P < .001). CONCLUSIONS Early-stage CRC patients can have distinct genomic profiles and CGP in this population can help expand access to targeted therapies.
Collapse
Affiliation(s)
- Leontios Pappas
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | | | | | - Aparna R Parikh
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
238
|
Meng X, Fan E, Lv D, Yang Y, Liu S. Associations between sleep traits and colorectal cancer: a mendelian randomization analysis. Front Oncol 2025; 15:1416243. [PMID: 39980544 PMCID: PMC11839420 DOI: 10.3389/fonc.2025.1416243] [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: 04/12/2024] [Accepted: 01/14/2025] [Indexed: 02/22/2025] Open
Abstract
Background Although many researches have shown a relationship between sleeping habits and the risk of developing colorectal cancer (CRC), there is a lack of data from randomized controlled trials (RCTs) to support this point. Hence, this study used Mendelian randomization (MR) to robustly assess whether five primary sleep characteristics are directly linked with the risk of CRC occurrence. Methods In the performed study, the main Mendelian randomization analysis was conducted using approaches such as Inverse Variance Weighting (IVW), MR Egger, and weighted median method. To this end, five genetically independent variants associated with the sleep-related characteristics (chronotype, sleep duration, insomnia, daytime napping, and daytime fatigue) were identified and used as instrumental variables. Publicly accessible GWAS (Genome-Wide Association Study) data were used to identify these variants to investigate the putative causal relationships between sleep traits and CRC. Additionally, we conducted sensitivity analyses to minimize possible biases and verify the consistency of our results. Results Mendelian randomization analyses showed that an morning chronotype reduces the risk of CRC with the IVW method, hence, odds ratio (OR) of 1.21 and 95% confidence interval (CI) of 0.67-0.93, which is statistically significant at P = 5.74E-03. Conversely, no significant evidence was found to suggest that sleep duration, insomnia, daytime napping, or daytime sleepiness have a direct causal impact on CRC risk according to the IVW analysis. Conclusions Findings from our Mendelian randomization analyses suggest that an individual's chronotype may contribute to an increased risk of CRC. It is advisable for individuals to adjust their sleep patterns as a preventative measure against CRC.
Collapse
Affiliation(s)
- Xiangyue Meng
- Department of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Enshuo Fan
- Department of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Dan Lv
- Department of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yongjing Yang
- Department of Radiotherapy, Jilin Cancer Hospital, Changchun, China
| | - Shixin Liu
- Department of Radiotherapy, Jilin Cancer Hospital, Changchun, China
| |
Collapse
|
239
|
Dey S, Ghosh M, Dev A. Signalling and molecular pathways, overexpressed receptors of colorectal cancer and effective therapeutic targeting using biogenic silver nanoparticles. Gene 2025; 936:149099. [PMID: 39557372 DOI: 10.1016/j.gene.2024.149099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/18/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Increasing morbidity and mortality in CRC is a potential threat to human health. The major challenges for better treatment outcomes are the heterogeneity of CRC cases, complicated molecular pathway cross-talks, the influence of gut dysbiosis in CRC, and the lack of multimodal target-specific drug delivery. The overexpression of many receptors in CRC cells may pave the path for targeting them with multiple ligands. The design of a more target-specific drug-delivery device with multiple ligand-functionalized, green-synthesized silver nanoparticles is highly promising and may also deliver other approved chemotherapeutic agents. This review presents the various aspects of colorectal cancer and over-expressed receptors that can be targeted with appropriate ligands to enhance the specific drug delivery potency of green synthesised silver nanoparticles. This review aims to broaden further research into this multi-ligand functionalised, safer and effective silver nano drug delivery system.
Collapse
Affiliation(s)
- Sandip Dey
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Manik Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Abhimanyu Dev
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India.
| |
Collapse
|
240
|
Lin C, Xu Z, XinLiang, Wei H, Wang X. A randomized controlled trial to compare the effect of oxycodone and sufentanil on postoperative analgesia and immune function for laparoscopic resection of colorectal cancer. BMC Anesthesiol 2025; 25:58. [PMID: 39910433 PMCID: PMC11796073 DOI: 10.1186/s12871-025-02922-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/21/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND The purpose of this study is to evaluate the effect of oxycodone and sufentanil on postoperative analgesia and immune function in patients with laparoscopic resection of colorectal cancer (CRC), as well as the serum level of inflammatory cytokine. METHODS 40 patients from August 2023 to August 2024 in Shenzhen Nanshan Hospital undergoing laparoscopic resection of CRC were randomly divided into Group O (n = 20) and Group S (n = 20). The visual analog scale (VAS) score and serial blood samples were assessed during perioperative period. The primary outcomes were VAS scores and immune indicators (including IL-2, C3, C4, IgG, IgA, IgE, IgM, CD3+, CD4+, CD8+ and CD4+/CD8+) at 24 h and 72 h post-surgery at 24 h and 72 h after surgery. The secondary outcomes were inflammatory markers (including IL-4, IL-6, IL-10, TNF-a and INF-y) at 24 h and 72 h after surgery. RESULTS The VAS scores at cough in Group O at 24 h and 72 h postoperative were lower than those in Group S (p < 0.001). No significant difference was found in VAS scores at rest between the two groups (P > 0.05). The immune indicators did not show significant changes after using oxycodone or sufentanil for patient-controlled intravenous analgesia (PCIA), respectively. There was no significant difference in inflammatory factors at 24 h and 72 h after surgery between the Group O and Group S. CONCLUSION Oxycodone is more effective than sufentanil in alleviating visceral pain, although it does not surpass sufentanil in managing cutting pain. In addition, there is no significant superiority in the effects of oxycodone on immune function and inflammatory cytokine release compared to sufentanil. TRIAL REGISTRATION Chinese Clinical Trial Registry (ChiCTR2400089072).
Collapse
Affiliation(s)
- Chunmei Lin
- The First Clinical Medical School, Jinan University, Guangzhou, 510632, People's Republic of China
- Department of Anaesthesiology, Shenzhen Nanshan Hospital, Shenzhen, 518000, People's Republic of China
| | - Zhiqiao Xu
- Department of Anaesthesiology, Shenzhen Nanshan Hospital, Shenzhen, 518000, People's Republic of China
| | - XinLiang
- Department of Anaesthesiology, Shenzhen Nanshan Hospital, Shenzhen, 518000, People's Republic of China
| | - Hong Wei
- Department of Anaesthesiology, Shenzhen Nanshan Hospital, Shenzhen, 518000, People's Republic of China.
| | - Xiaoping Wang
- Department of Pain Management, the First Affiliated Hospital of Jinan University, Guangzhou, 510632, People's Republic of China
| |
Collapse
|
241
|
Akkus E, Öksüz NE, Utkan G. Low-Density Lipoprotein (LDL) is Associated with Earlier Progression in Synchronous Metastatic Colorectal Cancer Treated without Curative Intent. J Gastrointest Cancer 2025; 56:58. [PMID: 39907838 DOI: 10.1007/s12029-025-01166-3] [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] [Accepted: 01/02/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Low-density lipoprotein cholesterol (LDL) is associated with the occurrence of colorectal cancer (CRC). This study aims to investigate its prognostic role and associated clinicopathological factors in the metastatic setting. METHODS Patients with newly diagnosed synchronous metastatic CRC were included. Patients were grouped according to the serum LDL levels at the diagnosis (≤ 130 mg/dL: Normal-LDL, > 130 mg/dL: High-LDL). LDL-associated clinicopathological factors, progression-free survival (PFS), and overall survival (OS) were assessed. RESULTS A total of 90 patients were included. 44.4% (n = 40) was in the normal-LDL and 56.6% (n = 50) in the high-LDL group. Colonic localization of the primary tumor was more frequent in the high-LDL group (90% vs. 67.5%, p = 0.009). The high-LDL group more frequently had local treatments [metastasectomy (26% vs. 2.5%, p = 0.002) and embolization-ablation (38% vs. 17.5%, p = 0.033)]. Despite higher curative intent with local treatments in the high-LDL group, PFS [10.03 months (95% Confidence Interval (CI):6.97-14.77) vs 9.63 mo. (95% CI: 7.93-14.00), p = 0.872] and OS [20.87 mo. (95% CI: 14.87-36.47) vs. 17.63 mo. (95% CI: 14.30-43.03), p = 0.925] did not differ from the normal-LDL. Among patients treated without any curative intent, high LDL was associated with significantly worse PFS [4.97 mo. (95% CI: 3.00-7.73) vs. 8.43 mo. (95% CI: 6.10-9.90), p = 0.048]. CONCLUSION This study suggests that serum LDL is associated with colonic primary localization in synchronous metastatic CRC. Levels > 130 mg/dL at diagnosis may be associated with worse survival and may be further investigated as a biomarker. Larger, multicenter and prospective studies are needed.
Collapse
Affiliation(s)
- Erman Akkus
- Ankara University Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye
- Ankara University Cancer Research Institute, Ankara, Türkiye
| | - Nejat Emre Öksüz
- Ankara University Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye
- Ankara University Cancer Research Institute, Ankara, Türkiye
| | - Güngör Utkan
- Ankara University Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye.
- Ankara University Cancer Research Institute, Ankara, Türkiye.
| |
Collapse
|
242
|
Kossenas K, Moutzouri O, Georgopoulos F. Comparison of short-term outcomes of robotic versus laparoscopic right colectomy for patients ≥ 65 years of age: a systematic review and meta-analysis of prospective studies. J Robot Surg 2025; 19:60. [PMID: 39904868 DOI: 10.1007/s11701-025-02222-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
Robotic right colectomy (RRC) has been researched and compared in the past to the laparoscopic approach (LRC). However, it remains unclear whether RRC outperforms LRC in patients aged 65 or older with colon cancer or adenoma. Thus, this systematic review and meta-analysis aims to compare the short-term outcomes of RRC to LRC in this particular age group. PubMed, Scopus, and Cochrane Library were searched for related literature. Studies and data were extracted by two independent reviewers. Inverse variances weighted mean differences (WMD) with random effects model were used for continuous values, and odds ratios (OR) with random effects model using the Mantel-Haenszel's formula were used for dichotomous values. Heterogeneity using Higgins I2 and p values were calculated. A sensitivity analysis was performed for operative duration and number of harvested lymph nodes. In total 382 patients, 157 in RRC and 225 in LRC, were included in this study. A statistically significant increase of 43.91 min [95%CI: 19.61, 68.22], P = 0.001, was observed regarding operative duration in the RRC group, compared to LRC, but with high heterogeneity, I2 = 89%. However, a statistically significant decrease of 42% [OR = 0.58 (0.34, 0.98), P = 0.04, I2=2%] was observed with regard to overall complications in RRC compared to LRC. Non-significant differences between the two approaches were observed in the number of harvested lymph nodes [WMD = 0.44 (95%CI: -3.94, 4.82), P = 0.85, I2 = 52%], wound infections [OR = 0.63 (95%CI: 0.11, 3.52), P = 0.60, I2 = 13%], rate of ileus [OR = 0.29 (95%CI: 0.08, 1.00), P = 0.05, I2 = 0%], length of hospitalization [WMD = 0.18 (95%CI: - 0.74, 1.11), P = 0.70, I2 = 0%], and anastomotic leakage [OR = 0.52 (95%CI: 0.09, 3.11), P = 0.47, I2 = 0%]. The results of the operative duration and number of harvested lymph nodes remained statistically significant and non-significant, respectively, after sensitivity analysis. Robotic right colectomy appears to require a longer operative duration, but possibly offers lower rates of overall complications, compared to laparoscopic right colectomy in patients ≥ 65 years of age. Due to the lack of studies identified in the literature, and the ones included being non-randomized, no solid conclusions can be drawn and cautious interpretation of the results is advised. Future studies are necessary to further examine both short- and long-term outcomes. Prospero registration: CRD42024603354.
Collapse
Affiliation(s)
- Konstantinos Kossenas
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, 21 Ilia Papakyriakou, 2414 Engomi, P.O. Box 24005, 1700, Nicosia, Cyprus.
| | - Olga Moutzouri
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, 21 Ilia Papakyriakou, 2414 Engomi, P.O. Box 24005, 1700, Nicosia, Cyprus
| | - Filippos Georgopoulos
- Head of Interventional Gastroenterology and Hepatology, Al Zahra Hospital, Dubai, UAE
| |
Collapse
|
243
|
Zhang G, Wu X, Fu H, Sun D. Circular RNA microarray expression profile and potential function of circDOCK1 in colorectal cancer. Front Genet 2025; 16:1443876. [PMID: 39967686 PMCID: PMC11832710 DOI: 10.3389/fgene.2025.1443876] [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: 06/04/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction Endoscopic tissue biopsy combined with histopathology is the gold standard for the diagnosis of colorectal cancer (CRC); however, the invasive nature of this procedure hinders its acceptance by patients. Therefore, there exists a critical need to identify novel markers facilitating early CRC detection and prognosis. Circular RNAs (circRNAs) hold promise as novel clinical diagnostic markers. This study aimed to investigate the impact of circDOCK1 on CRC metastasis and prognosis as well as its underlying molecular mechanisms. Methods We explored circRNA expression profiles in four pairs of CRC tissues and adjacent non-carcinoma tissues via microarray analysis. After Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and circRNA-miRNA network analyses, circDOCK1 was chosen for further investigation. We evaluated its clinical relevance in 80 CRC tissue pairs and adjacent controls, correlating circDOCK1 expression with clinical characteristics. Follow-up data from patient telephone interviews were analyzed for survival outcomes. Transfection efficiency was confirmed via qRT-PCR in HCT116 and SW480 colon cells, and the effects of circDOCK1 on cell proliferation, migration, and invasion were assessed. Results Microarray data revealed 149 significantly differentially expressed circRNAs, including 71 upregulated and 78 downregulated circRNAs, in CRC tissues. CircDOCK1 exhibited elevated expression in patients with CRC and emerged as an independent prognostic factor. Kaplan-Meier curve analysis suggested that circDOCK1 expression is an unfavorable prognostic factor in patients with CRC. In vivo experiments revealed that circDOCK1 overexpression enhanced the proliferation, migration, and invasion of CRC cells, with consistent results upon circDOCK1 downregulation. Conclusion These data indicate that circDOCK1 may play a role in promoting the proliferation, migration, and invasion of CRC cells, suggesting its potential as a CRC biomarker.
Collapse
Affiliation(s)
| | | | | | - Daqing Sun
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
244
|
Kou P, Lin L, Li Y, Qin H, Zhang K, Zhang W, Li J, Zhang Y, Cheng J. Application of cellular microstructural diffusion MRI (cell size imaging) in rectal lesions: a preliminary study. Front Oncol 2025; 15:1535271. [PMID: 39963105 PMCID: PMC11830574 DOI: 10.3389/fonc.2025.1535271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Objectives To explore the value of cellular microstructural mapping by IMPULSED (imaging microstructural parameters using limited spectrally edited diffusion) method in evaluating the histological type and prognostic factors of rectal lesions. Materials and methods Sixty-six patients with rectal lesions were enrolled in this study. All subjects underwent MRI scans including conventional diffusion weighted imaging (DWI) and the IMPULSED MRI scans of oscillating gradient spin-echo (OGSE) and pulse gradient spin-echo (PGSE) sequences. Parameters including mean cell diameter (dmean), intracellular fraction (vin), extracellular diffusivity (dex), cellularity, and apparent diffusion coefficient (ADC) values (ADCPGSE, ADC17Hz, ADC33Hz, and ADC of conventional DWI) were measured in different histopathologic types, grades, stages, and structure invasion statuses. The receiver operating characteristic (ROC) curve analysis was used to evaluate diagnostic power. The sensitivity, specificity, and the corresponding area under the curves (AUCs) were calculated. Results Our preliminary results illustrated that malignant lesion showed higher vin and cellularity ([0.2867 ± 0.0697] vs. [0.1856 ± 0.1011], [2.3508 ± 0.6055] vs. [1.2716 ± 0.4574], all P<0.05), lower dex and ADC values (ADCPGSE, ADC17Hz, and ADC of conventional DWI) compared to benign lesion ([2.1637 ± 0.3303 μm2/ms] vs. [2.5595 ± 0.5085 μm2/ms], [0.9238 (0.7959, 1.0741) ×10-3 mm2/s] vs. [1.3373 ± 0.3902×10-3 mm2/s], [1.3204 ± 0.2342×10-3 mm2/s] vs. [1.8029 ± 0.3119×10-3 mm2/s], [0.7400 (0.6750, 0.8375) ×10-3 mm2/s] vs. [1.0550 ± 1.1191×10-3 mm2/s], all P<0.05), while no significant difference was seen for dmean. Vin and cellularity of rectal common adenocarcinoma (AC) were significantly higher than those of rectal mucinous adenocarcinoma (MC) ([0.2994 ± 0.0626] vs. [0.2028 ± 0.0571], [2.4579 ± 0.5553] vs. [1.6412 ± 0.4347], all P<0.05), while dex and ADC values (ADCPGSE, ADC17Hz, ADC33Hz, and ADC of conventional DWI) were lower in AC ([2.1189 ± 0.3187 μm2/ms] vs. [2.4609 ± 0.2534 μm2/ms], [0.8996 ± 0.1583×10-3 mm2/s] vs. [1.2072 ± 0.2326×10-3 mm2/s], [1.2714 ± 0.1916×10-3 mm2/s] vs. [1.6451 ± 0.2420×10-3 mm2/s], [1.8963 (1.6481, 2.1138) ×10-3 mm2/s] vs. [2.3104 ± 0.3851×10-3 mm2/s], [0.7341 ± 0.8872×10-3 mm2/s] vs. [1.1410 ± 0.1840×10-3 mm2/s], all P<0.05). In AC group, the dmean had significant difference between negative and positive tumor budding (TB) ([13.2590 ± 1.3255 μm] vs. [14.3014 ± 1.1830 μm], P<0.05). No significant difference of dmean, vin, dex, cellularity or ADC values was observed in AC with different grade, T stage, N stage, perineural and lymphovascular invasion (all P>0.05). The ROC curves showed that the area under the curves (AUCs) of vin, dex, cellularity, and ADC values (ADCPGSE, ADC17Hz, and ADC of conventional DWI) for distinguishing malignant and benign lesion were 0.803, 0.757, 0.948, 0.807, 0.908 and 0.905, respectively. The AUCs of vin, dex, cellularity, and ADC values (ADCPGSE, ADC17Hz, ADC33Hz, and ADC of conventional DWI) in distinguishing AC from MC were 0.887, 0.802, 0.906, 0.896, 0.896, 0.781 and 0.991, respectively. The AUC of the dmean for evaluating TB status was 0.726. The AUC of ADC from conventional DWI for evaluating WHO grade was 0.739. Conclusion Cellular microstructural mapping by the IMPULSED method has great potential in preoperative evaluation of rectal lesions. It could be helpful in differentiating malignant and benign lesions, distinguishing AC from MC, and in predicting the TB status.
Collapse
Affiliation(s)
- Peisi Kou
- Department of Magnetic Resonance Imaging (MRI), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liangjie Lin
- Clinical and Technical Support, Philips Healthcare, Beijing, China
| | - Ying Li
- Department of Magnetic Resonance Imaging (MRI), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Qin
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Zhang
- Department of Magnetic Resonance Imaging (MRI), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenhua Zhang
- Department of Magnetic Resonance Imaging (MRI), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Juan Li
- Department of Magnetic Resonance Imaging (MRI), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yong Zhang
- Department of Magnetic Resonance Imaging (MRI), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging (MRI), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
245
|
Jian Y, Yang S, Liu R, Tan X, Zhao Q, Wu J, Chen Y. Radiomics Analysis of Different Machine Learning Models based on Multiparametric MRI to Identify Benign and Malignant Testicular Lesions. Acad Radiol 2025:S1076-6332(25)00067-4. [PMID: 39904666 DOI: 10.1016/j.acra.2025.01.026] [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: 12/05/2024] [Revised: 01/18/2025] [Accepted: 01/19/2025] [Indexed: 02/06/2025]
Abstract
RATIONALE AND OBJECTIVES To develop and validate a machine learning-based prediction model for the use of multiparametric magnetic resonance imaging(MRI) to predict benign and malignant lesions in the testis. MATERIALS AND METHODS The study retrospectively enrolled 148 patients with pathologically confirmed benign and malignant testicular lesions, dividing them into: training set (n=103) and validation set (n=45). Radiomics characteristics were derived from T2-weighted(T2WI)、contrast-enhanced T1-weighted(CE-T1WI)、diffusion-weighted imaging(DWI) and Apparent diffusion coefficient(ADC) MRI images, followed by feature selection. A machine learning-based combined model was developed by incorporating radiomics scores (rad scores) from the optimal radiomics model along with clinical predictors. Draw the receiver operating characteristic (ROC) curve and use the area under the curve (AUC) to evaluate and compare the predictive performance of each model. The diagnostic efficacy of the various machine learning models was evaluated using the Delong test. RESULTS Radiomics features were extracted from four sequence-based groups(CE-T1WI+DWI+ADC+T2WI), and the model that combined Logistic Regression(LR) machine learning showed the best performance in the radiomics model. The clinical model identified one independent predictors. The combined clinical-radiomics model showed the best performance, whose AUC value was 0.932(95% confidence intervals(CI)0.868-0.978), sensitivity was 0.875, specificity was 0.871 and accuracy was 0.884 in validation set. CONCLUSION The combined clinical-radiomics model can be used as a reliable tool to predict benign and malignant testicular lesions and provide a reference for clinical treatment method decisions.
Collapse
Affiliation(s)
- Yuanxi Jian
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China (Y.J., S.Y., R.L., X.T., Q.Z., J.W., Y.C.).
| | - Suping Yang
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China (Y.J., S.Y., R.L., X.T., Q.Z., J.W., Y.C.)
| | - Rui Liu
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China (Y.J., S.Y., R.L., X.T., Q.Z., J.W., Y.C.)
| | - Xin Tan
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China (Y.J., S.Y., R.L., X.T., Q.Z., J.W., Y.C.)
| | - Qian Zhao
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China (Y.J., S.Y., R.L., X.T., Q.Z., J.W., Y.C.)
| | - Junlin Wu
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China (Y.J., S.Y., R.L., X.T., Q.Z., J.W., Y.C.)
| | - Yuan Chen
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China (Y.J., S.Y., R.L., X.T., Q.Z., J.W., Y.C.)
| |
Collapse
|
246
|
Piercey O, Chantrill L, Hsu H, Ma B, Price T, Tan IB, Teng H, Tie J, Desai J. Expert consensus on the optimal management of BRAF V600E-mutant metastatic colorectal cancer in the Asia-Pacific region. Asia Pac J Clin Oncol 2025; 21:31-45. [PMID: 39456063 PMCID: PMC11733838 DOI: 10.1111/ajco.14132] [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: 05/15/2024] [Revised: 09/14/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024]
Abstract
The burden of colorectal cancer (CRC) is high in the Asia-Pacific region, and several countries in this region have among the highest and/or fastest growing rates of CRC in the world. A significant proportion of patients will present with or develop metastatic CRC (mCRC), and BRAFV600E-mutant mCRC represents a particularly aggressive phenotype that is less responsive to standard chemotherapies. In light of recent therapeutic advances, an Asia-Pacific expert consensus panel was convened to develop evidence-based recommendations for the diagnosis, treatment, and management of patients with BRAFV600E-mutant mCRC. The expert panel comprised nine medical oncologists from Australia, Hong Kong, Singapore, and Taiwan (the authors), who met to review current literature and develop eight consensus statements that describe the optimal management of BRAFV600E-mutant mCRC in the Asia-Pacific region. As agreed by the expert panel, the consensus statements recommend molecular testing at diagnosis to guide individualized treatment decisions, propose optimal treatment pathways according to microsatellite stability status, advocate for more frequent monitoring of BRAFV600E-mutant mCRC, and discuss local treatment strategies for oligometastatic disease. Together, these expert consensus statements are intended to optimize treatment and improve outcomes for patients with BRAFV600E-mutant mCRC in the Asia-Pacific region.
Collapse
Affiliation(s)
| | - Lorraine Chantrill
- Illawarra Shoalhaven Local Health DistrictIllawarraNew South WalesAustralia
- Faculty of Science, Medicine and HealthUniversity of WollongongWollongongNew South WalesAustralia
| | - Hung‐Chih Hsu
- Division of Hematology OncologyChang Gung Memorial HospitalNew TaipeiTaiwan
- College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Brigette Ma
- State Key Laboratory of Translational Oncology, Sir YK Pao Centre for Cancer, Department of Clinical Oncology, Hong Kong Cancer InstituteThe Chinese University of Hong KongHong Kong SARChina
| | - Timothy Price
- The Queen Elizabeth HospitalAdelaideSouth AustraliaAustralia
| | - Iain Beehuat Tan
- Division of Medical OncologyNational Cancer Centre SingaporeSingaporeSingapore
| | - Hao‐Wei Teng
- Department of OncologyTaipei Veterans General HospitalTaipeiTaiwan
| | - Jeanne Tie
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Jayesh Desai
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
247
|
Ishiyama Y, Hirano Y, Minagawa Y, Yamato M, Akuta S, Nakanishi A, Fujii T, Okazaki N, Hiranuma C, Koyama I. A Case of Transanal Total Mesorectal Excision Using the Senhance Digital Laparoscopy System for Rectal Cancer. Surg Case Rep 2025; 11:24-0024. [PMID: 39991492 PMCID: PMC11842874 DOI: 10.70352/scrj.cr.24-0024] [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: 10/11/2024] [Accepted: 12/09/2024] [Indexed: 02/25/2025] Open
Abstract
INTRODUCTION This case report presents the world's first transanal total mesorectal excision (taTME) using the Senhance Digital Laparoscopy System for rectal cancer. Senhance has been gaining attention as an advanced surgical robot, providing better visualization, stable dissection, and reduced surgeon fatigue compared to conventional methods. CASE PRESENTATION A 68-year-old woman underwent Senhance-assisted taTME for a rectal tumor located 4 cm from the anal verge. The procedure was completed safely with a total operation time of 209 minutes, a cockpit time of 85 minutes, and a blood loss of 40 mL. Pathological evaluation confirmed complete resection (R0) with no residual cancer or lymph node metastases. The patient was discharged on postoperative day 7 without complications. CONCLUSIONS This case demonstrates the potential advantages of Senhance-assisted taTME, including improved visualization, stable dissection, and reduced surgeon fatigue. Further studies are needed to evaluate long-term outcomes and establish the role of this technique in rectal cancer surgery.
Collapse
Affiliation(s)
- Yasuhiro Ishiyama
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| | - Yasumitsu Hirano
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| | - Yume Minagawa
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| | - Misuzu Yamato
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| | - Sohei Akuta
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| | - Akihito Nakanishi
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| | - Takatsugu Fujii
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| | - Naoto Okazaki
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| | - Chikashi Hiranuma
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| | - Isamu Koyama
- Department of Gastroenterological Surgery, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| |
Collapse
|
248
|
Amintas S, Dupin C, Derieppe MA, Moranvillier I, Lamrissi I, Bourdié C, Feurer Z, Fernandez B, Heng-Pradère T, Moreau-Gaudry F, Bedel A, Vendrely V, Dabernat S. Resveratrol and capsaicin as safer radiosensitizers for colorectal cancer compared to 5-fluorouracil. Biomed Pharmacother 2025; 183:117799. [PMID: 39755023 DOI: 10.1016/j.biopha.2024.117799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/14/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND AND AIM Standard rectal cancer treatment includes neoadjuvant radiotherapy sensitized by 5-fluorouracil (5-FU) chemotherapy. However, 5-FU increased chemoradiotherapy response rate comes with significant toxicity, especially in older, frail patients. The development of alternatives to chemotherapy enabling radiosensitization with limited systemic toxicity is therefore needed to improve patient management. Bioactive food components (BFCs) can exhibit chemo or radio-sensitizing properties against cancer cells. Moreover, the cytotoxic action of BFCs may be tumor-specific, with reduced impact on healthy cells. We hypothesized that BFCs, in particular resveratrol and capsaicin, alone or in association, could lead to specific radio-sensitization of colorectal tumors while offering reduced toxicity compared to 5-FU. EXPERIMENTAL PROCEDURE Colorectal tumor and non-tumor cell lines were treated with resveratrol, capsaicin, or 5-FU, alone or in combination, then irradiated; survival, cell cycle, and apoptosis were analyzed. RAGγ2C-/- mice with xenografts received oral resveratrol, resveratrol + capsaicin, or 5-FU, followed by radiotherapy, with tumor growth and systemic toxicity evaluated. KEY RESULTS Resveratrol alone or in association with capsaicin radio-potentiates colorectal tumor cells in vitro, impacting both cell cycle and apoptosis. In a preclinical mouse model, the oral administration of resveratrol and capsaicin, but not resveratrol alone, allowed the radio-sensitization of subcutaneous colorectal tumors with similar efficiency to 5-FU. Moreover, the global as well as the hematological toxicity of the BFC association was lower than those of 5-FU. CONCLUSION This work establishes BFCs as effective enhancers of radiotherapy, offering a safer alternative to traditional radiosensitization with chemotherapy.
Collapse
Affiliation(s)
- Samuel Amintas
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France; Tumor Biology and Tumor Bank Department, Bordeaux University Hospital, Bordeaux, France
| | - Charles Dupin
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France; Radiotherapy Department, Bordeaux University Hospital, Bordeaux, France
| | - Marie-Alix Derieppe
- University of Bordeaux, Bordeaux, France; Shared Animal Facility, Common Animal Facilities Service, University of Bordeaux, Bordeaux, France
| | - Isabelle Moranvillier
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France
| | - Isabelle Lamrissi
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France
| | - Corine Bourdié
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France
| | - Zoe Feurer
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France
| | - Benjamin Fernandez
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France; Colorectal Digestive Surgery Department, Bordeaux University Hospital (CHU de Bordeaux), France
| | - Tyty Heng-Pradère
- Pathology Department, Bordeaux University Hospital (CHU de Bordeaux), Bordeaux, France
| | - François Moreau-Gaudry
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France; Biochemistry Department, Bordeaux University Hospital (CHU de Bordeaux), Bordeaux, France
| | - Aurélie Bedel
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France; Biochemistry Department, Bordeaux University Hospital (CHU de Bordeaux), Bordeaux, France
| | - Véronique Vendrely
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France; Radiotherapy Department, Bordeaux University Hospital, Bordeaux, France
| | - Sandrine Dabernat
- University of Bordeaux, Bordeaux, France; INSERM U1312, Bordeaux Institute of Oncology - BRIC, BioGo Team, Bordeaux, France; Biochemistry Department, Bordeaux University Hospital (CHU de Bordeaux), Bordeaux, France.
| |
Collapse
|
249
|
Gila F, Khoddam S, Jamali Z, Ghasemian M, Shakeri S, Dehghan Z, Fallahi J. Personalized medicine in colorectal cancer: a comprehensive study of precision diagnosis and treatment. Per Med 2025; 22:59-81. [PMID: 39924822 DOI: 10.1080/17410541.2025.2459050] [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: 07/30/2024] [Accepted: 01/23/2025] [Indexed: 02/11/2025]
Abstract
Colorectal cancer is a common and fatal disease that affects many people globally. CRC is classified as the third most prevalent cancer among males and the second most frequent cancer among females worldwide. The purpose of this article is to examine how personalized medicine might be used to treat colorectal cancer. The classification of colorectal cancer based on molecular profiling, including the detection of significant gene mutations, genomic instability, and gene dysregulation, is the main topic of this discussion. Advanced technologies and biomarkers are among the detection methods that are explored, demonstrating their potential for early diagnosis and precise prognosis. In addition, the essay explores the world of treatment possibilities by providing light on FDA-approved personalized medicine solutions that provide individualized and precise interventions based on patient characteristics. This article assesses targeted treatments like cetuximab and nivolumab, looks at the therapeutic usefulness of biomarkers like microsatellite instability (MSI) and circulating tumor DNA (ctDNA), and investigates new approaches to combat resistance. Through this, our review provides a thorough overview of personalized medicine in the context of colorectal cancer, ultimately highlighting its potential to revolutionize the field and improve patient care.
Collapse
Affiliation(s)
- Fatemeh Gila
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Khoddam
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Jamali
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohmmad Ghasemian
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shayan Shakeri
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zeinab Dehghan
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
250
|
Goldberg SR, Ko LK, Hsu L, Yin H, Kooperberg C, Peters U, Burnett-Hartman AN. Patient Perspectives on Personalized Risk Communication Using Polygenic Risk Scores to Inform Colorectal Cancer Screening Decisions. AJPM FOCUS 2025; 4:100308. [PMID: 39866161 PMCID: PMC11761838 DOI: 10.1016/j.focus.2024.100308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Introduction Colorectal cancer is increasingly diagnosed in people aged <50 years. New U.S. guidelines recommend screening initiation at age 45 years. Providing personalized risk for colorectal cancer using polygenic risk scores may be an opportunity to engage this younger population in colorectal cancer screening. There is limited research on patient understanding of polygenic risk scores results and use of polygenic risk scores to inform colorectal cancer screening decisions. Methods From May 2022 to June 2023, 20 Kaiser Permanente Colorado members aged 46-51 years who had been offered colorectal cancer screening but had never completed it signed consent to provide a saliva sample for colorectal cancer polygenic risk score analysis. After receiving personalized polygenic risk scores for colorectal cancer, participants completed a semistructured interview regarding the understanding of their polygenic risk scores, perceived colorectal cancer risk, and intention to screen. Thematic analysis was conducted using Atlas.ti, Version 8. Results Of the 19 participants who successfully completed polygenic risk score-related testing and a semistructured interview, 13 were female, 14 never smoked cigarettes, 6 were Hispanic, and 13 were non-Hispanic White. One participant had high risk for colorectal cancer on the basis of polygenic risk score results. Qualitative interviews showed participants' understanding of their results, trust in polygenic risk scores, perception of risk for colorectal cancer, plans to complete colorectal cancer screening, intent to share polygenic risk scores with healthcare providers, and concerns about genetic results impacting health care. Conclusions Qualitative analyses suggest that participants were interested in and understood their polygenic risk score results. Further study is needed to develop guidelines, effective calls to action, provider engagement, and health education materials on use of polygenic risk scores for health decision making.
Collapse
Affiliation(s)
- Shauna R. Goldberg
- Kaiser Permanente Colorado Institute for Health Research, Aurora, Colorado
| | - Linda K. Ko
- Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Health Systems and Population Health, University of Washington, Seattle, Washington
| | - Li Hsu
- Fred Hutchinson Cancer Center, Seattle, Washington
| | - Hang Yin
- Fred Hutchinson Cancer Center, Seattle, Washington
| | | | | | - Andrea N. Burnett-Hartman
- Kaiser Permanente Colorado Institute for Health Research, Aurora, Colorado
- Fred Hutchinson Cancer Center, Seattle, Washington
| |
Collapse
|