1
|
Sultan MH, Zhan Q, Wang Y, Xia Y, Jia X. Precision oncolytic viral therapy in colorectal cancer: Genetic targeting and immune modulation for personalized treatment (Review). Int J Mol Med 2025; 56:104. [PMID: 40342021 PMCID: PMC12081034 DOI: 10.3892/ijmm.2025.5545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/09/2025] [Indexed: 05/11/2025] Open
Abstract
Colorectal cancer (CRC) is a leading health issue and treatments to eradicate it, such as conventional chemotherapy, are non‑selective and come with a number of complications. The present review focuses on the relatively new area of precision oncolytic viral therapy (OVT), with genetic targeting and immune modifications that offer a new future for CRC treatment. In the present review, an overview of the selection factors that are considered optimal for an oncolytic virus, mechanisms of oncolysis and immunomodulation applied to the OVT, as well as new strategies to improve the efficacy of this method are described. Additionally, cause‑and‑effect relationships are examined for OVT efficacy, mediated by the tumor microenvironment, and directions for genetic manipulation of viral specificity are explored. The possibility of synergy between OVT and immune checkpoint inhibitors and other treatment approaches are demonstrated. Incorporating the details of the present review, biomarker‑guided combination therapies in precision OVT for individualized CRC care, significant issues and future trends in this required area of medicine are highlighted. Increasingly, OVT is leaving the experimental stage and may become routine practice; it provides a new perspective on overcoming CRC and highlights the importance of further research and clinical work.
Collapse
Affiliation(s)
- Muhammad Haris Sultan
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
- Center for Translational Medicine and Precision Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, P.R. China
| | - Qi Zhan
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Yigang Wang
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Yulong Xia
- Center for Translational Medicine and Precision Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiaoyuan Jia
- College of Life Sciences and Medicine, Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| |
Collapse
|
2
|
Li M, Gao X, Lin X, Zhang Y, Peng W, Sun T, Shu W, Shi Y, Guan Y, Xia X, Yi X, Li Y, Jia J. Analysis of germline-somatic mutational connections in colorectal cancer reveals differential tumorigenic patterns and a novel predictive marker for germline mutation carriers. Cancer Lett 2025; 620:217637. [PMID: 40118241 DOI: 10.1016/j.canlet.2025.217637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/23/2025]
Abstract
Colorectal cancer (CRC) genetic testing of regions beyond clinical guidelines has revealed a substantial number of likely pathogenic germline mutations (GMs). It remains largely undetermined whether and how these GMs, typically located in non-mismatch repair (non-MMR) genes, are associated with the tumorigenesis of CRC. This study aimed to identify CRC-predisposing GMs among 93 cancer susceptibility genes and investigate their potential influences on CRC somatic mutational features. We secondarily aimed to investigate whether somatic ERBB2 amplification contributes to identifying GM carriers. This study incorporated a total of 3,240 Chinese CRC patients and 10,588 control individuals. CRC patients were subjected to paired tumor-normal sequencing with a 1,021-gene panel. A case-control analysis was conducted to profile the GM-associated CRC risk. A comprehensive germline-somatic association analysis was performed among 2,405 patients, with key findings subsequently validated in an independent 835-patient cohort and the TCGA CRC cohort. The case-control results supported CRC-predisposing effects of GMs in certain homologous recombination repair (HRR) and DNA damage checkpoint factor (CPF) genes, such as BRCA1/2, RecQ helicase genes, ATM, and CHEK2. HRR GMs were associated with an increased copy number alteration burden, more TP53 clonal mutations, and a higher probability of carrying somatic ERBB2 amplification. CPF GMs were inferred to have synergistic effects with ARID1A and KDM6A somatic mutations in CRC tumorigenesis. Among patients with onset age ≥55 years, stable microsatellites, and no cancer family history, ERBB2 amplification was significantly predictive of GM carriers. Our findings elucidate different germline tumorigenic patterns not driven by deficient MMR. Somatic ERBB2 amplification in CRC can serve as an indicator for germline genetic testing when traditional risk features are absent.
Collapse
Affiliation(s)
- Mintao Li
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Xuan Gao
- Geneplus-Shenzhen Clinical Laboratory, Shenzhen, China
| | - Xiangchun Lin
- Department of Gastroenterology, Peking University International Hospital, Beijing, China
| | - Yan Zhang
- Geneplus-Beijing Institute, Beijing, China
| | - Wenying Peng
- The Second Department of Oncology, Yunnan Cancer Hospital & the Third Affiliated Hospital of Kunming Medical University & Yunnan Cancer Center, Kunming, China
| | - Tao Sun
- General Surgery Department, Peking University Third Hospital, Beijing, China
| | - Weiyang Shu
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Yanyan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | | | | | - Xin Yi
- Geneplus-Beijing Institute, Beijing, China.
| | - Yuan Li
- Department of Gastroenterology, Peking University International Hospital, Beijing, China; Department of Gastroenterology, Peking University Third Hospital, Beijing, China.
| | - Jinzhu Jia
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China; Center for Statistical Science, Peking University, Beijing, China.
| |
Collapse
|
3
|
Viot J, Loyon R, Adib N, Laurent-Puig P, de Reyniès A, André F, Monnien F, André T, Svrcek M, Turpin A, Selmani Z, Arnould L, Guyard L, Gilbert N, Boureux A, Adotevi O, Vienot A, Abdeljaoued S, Vernerey D, Borg C, Gautheret D. Deciphering human endogenous retrovirus expression in colorectal cancers: exploratory analysis regarding prognostic value in liver metastases. EBioMedicine 2025; 116:105727. [PMID: 40381378 DOI: 10.1016/j.ebiom.2025.105727] [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: 08/09/2024] [Revised: 04/02/2025] [Accepted: 04/12/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Human Endogenous RetroVirus (HERV) expression in tumours reflects epigenetic dysregulation of cancer and an oncogenic factor through promoter/enhancer action on genes. While more than 50% of colorectal cancers develop liver metastases, HERV has not been studied in this context. METHODS We collected 400 RNA-seq samples from over 200 patients with primary and liver metastases, including public data and a novel set of 200 samples. FINDINGS We observed global stability of HERV expression between liver metastases and primary colorectal cancers, suggesting an early oncogenic footprint. We identified a list of 17 HERV loci for liver metastatic colorectal cancer (lmCRC) characterization; with tumour-specificity validated in single-cell metastatic colorectal cancer data and normal tissue bulk RNA-seq. Eleven loci produced HERV-derived peptides as per tandem mass spectrometry from primary colorectal cancer. Six loci were associated with the risk of relapse after lmCRC surgery. Four, HERVH_Xp22.32a, HERVH_20p11.23b, HERVH_13q33.3, HERVH_13q31.3, had adverse prognostic value (log-rank p-value 0.028, 0.0083, 9e-4, 0.05, respectively) while two, HERVH_Xp22.2c (log-rank p-value 0.032) and HERVH_8q21.3b (in multivariable models) were associated with better prognosis. Moreover, the markers showed a cumulative effect on survival when expressed. Some were associated with decreased cytotoxic immune cells and most of them correlated with cell cycle pathways. INTERPRETATION These findings provide insights into the lmCRC transcriptome landscape by suggesting prognostic markers and potential therapeutic targets. FUNDING This work was supported by funding from institutional grants from Inserm, EFS, University of Bourgogne Franche-Comté, national found "Agence Nationale de la Recherche - ANR-JCJC: Projet HERIC and ANR-22-CE45-0007", and "La ligue contre le cancer".
Collapse
Affiliation(s)
- Julien Viot
- Département d'Oncologie Médicale, CHU Besançon, Besançon 25000, France; Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.
| | - Romain Loyon
- Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Nawfel Adib
- Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Pierre Laurent-Puig
- Department of Biology, Institut du Cancer Paris CARPEM, APHP, APHP.Centre-Université Paris Cité, Hôpital Européen G. Pompidou, Paris, France; Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, EPIGENETEC, Paris 75006, France
| | - Aurélien de Reyniès
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, EPIGENETEC, Paris 75006, France
| | - Fabrice André
- Paris-Saclay University, Gustave Roussy, Villejuif, France; Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Franck Monnien
- Département d'Oncologie Médicale, CHU Besançon, Besançon 25000, France; Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Thierry André
- Department of Medical Oncology, Sorbonne University, Saint-Antoine Hospital, AP-HP, Paris, France
| | - Magali Svrcek
- Department of Pathology, Saint-Antoine Hospital, AP-HP, Sorbonne Université, Paris, France
| | - Anthony Turpin
- Department of Oncology, Lille University Hospital, France; CNRS UMR9020, INSERM UMR1277, University of Lille, Institut Pasteur, Lille, France
| | - Zohair Selmani
- Département d'Oncologie Médicale, CHU Besançon, Besançon 25000, France; Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Laurent Arnould
- Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, Dijon, France; CCRB Ferdinand Cabanne de Dijon, France
| | - Laura Guyard
- Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, Dijon, France; CCRB Ferdinand Cabanne de Dijon, France
| | - Nicolas Gilbert
- IRMB, INSERM U1183, Hopital Saint-Eloi, Universite de Montpellier, Montpellier, France
| | - Anthony Boureux
- IRMB, INSERM U1183, Hopital Saint-Eloi, Universite de Montpellier, Montpellier, France
| | - Olivier Adotevi
- Département d'Oncologie Médicale, CHU Besançon, Besançon 25000, France; Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Angélique Vienot
- Département d'Oncologie Médicale, CHU Besançon, Besançon 25000, France; Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Syrine Abdeljaoued
- Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Dewi Vernerey
- Département d'Oncologie Médicale, CHU Besançon, Besançon 25000, France
| | - Christophe Borg
- Département d'Oncologie Médicale, CHU Besançon, Besançon 25000, France; Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Daniel Gautheret
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CNRS, CEA, Gif-sur-Yvette 91190, France
| |
Collapse
|
4
|
Wang LJ, Bao Q, Wang HW, Huang LF, Zhang JQ, Zhao TT, Jin KM, Liu XF, Wang K, Li ZW, Xing BC. Predictive Factors for Chemotherapy Response in Colorectal Liver Metastasis: A Retrospective Study Utilizing Next-Generation Sequencing. Ann Surg Oncol 2025:10.1245/s10434-025-17320-x. [PMID: 40342004 DOI: 10.1245/s10434-025-17320-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/30/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND This study aimed to identify predictive factors for chemotherapy response in colorectal liver metastasis (CRLM) patients. METHODS Eligible participants with CRLM who had undergone at least two systemic chemotherapy cycles postdiagnosis were retrospectively analyzed. They were categorized as responders and nonresponders based on tumor size reduction. DNA extracted from tumor tissues was subjected to sequencing. Additionally, a comparative analysis of oncogenic pathways was conducted. Logistic regression analysis was conducted to determine predictive factors for chemotherapy response. RESULTS A total of 230 Chinese patients were analyzed. Significant differences in mutation distribution were found, particularly in the KRAS gene and several specific rare gene mutations (EP300, PTPRK, KMT2A, and ACVR1B), as well as in the PI3K and RTK-RAS pathways between the two groups. Gender, utilization of biological targeted agents (BTAs), KRAS gene mutations, PI3K pathway alterations, and specific rare gene mutations were used to construct a specific efficacy prediction model, achieving an area under the curve (AUC) of 0.73. Approximately 75% (87/116) of patients could potentially avoid BTAs based on the model's predictions. In a subgroup of 52 patients not using BTAs, simulation indicated that 10 patients could benefit by including BTAs, representing 32% (10 of 31) of initially nonresponsive patients. CONCLUSIONS Gender, utilization of BTAs, and specific gene and pathway mutations may be significant predictors of chemotherapy response in CRLM patients. These findings highlight the role of genetic profiling in refining treatment strategies.
Collapse
Affiliation(s)
- Li-Jun Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery Unit I, Peking University Cancer Hospital and Institute, Beijing, China
| | - Quan Bao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery Unit I, Peking University Cancer Hospital and Institute, Beijing, China
| | - Hong-Wei Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery Unit I, Peking University Cancer Hospital and Institute, Beijing, China
| | - Long-Fei Huang
- GloriousMed Clinical Laboratory Co., Ltd., Research Institute, Shanghai, China
| | - Jing-Qing Zhang
- GloriousMed Clinical Laboratory Co., Ltd., Research Institute, Shanghai, China
| | - Ting-Ting Zhao
- GloriousMed Clinical Laboratory Co., Ltd., Research Institute, Shanghai, China
| | - Ke-Min Jin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery Unit I, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiao-Feng Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery Unit I, Peking University Cancer Hospital and Institute, Beijing, China
| | - Kun Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery Unit I, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhong-Wu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China.
| | - Bao-Cai Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery Unit I, Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
5
|
Emami A, Mahdavi Sharif P, Rezaei N. KRAS mutations in colorectal cancer: impacts on tumor microenvironment and therapeutic implications. Expert Opin Ther Targets 2025:1-23. [PMID: 40320681 DOI: 10.1080/14728222.2025.2500426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/24/2025] [Indexed: 05/08/2025]
Abstract
INTRODUCTION Despite decreasing trends in incidence, colorectal cancer (CRC) is still a major contributor to malignancy-related morbidities and mortalities. Groundbreaking advances in immunotherapies and targeted therapies benefit a subset of CRC patients, with sub-optimal outcomes. Hence, there is an unmet need to design and manufacture novel therapies, especially for advanced/metastatic disease. KRAS, the most highly mutated proto-oncogene across human malignancies, particularly in pancreatic adenocarcinoma, non-small cell lung cancer, and CRC, is an on-off switch and governs several fundamental cell signaling cascades. KRAS mutations not only propel the progression and metastasis of CRC but also critically modulate responses to targeted therapies. AREAS COVERED We discuss the impacts of KRAS mutations on the CRC's tumor microenvironment and describe novel strategies for targeting KRAS and its associated signaling cascades and mechanisms of drug resistance. EXPERT OPINION Drug development against KRAS mutations has been challenging, mainly due to structural properties (offering no appropriate binding site for small molecules), critical functions of the wild-type KRAS in non-cancerous cells, and the complex network of its downstream effector pathways (allowing malignant cells to develop resistance). Pre-clinical and early clinical data offer promises for combining KRAS inhibitors with immunotherapies and targeted therapies.
Collapse
Affiliation(s)
- Anita Emami
- Tehran University of Medical Sciences, Tehran, Iran
| | | | - Nima Rezaei
- Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Zhang L, Huang Q, Yang J, Hu Y. Bardoxolone methyl suppressed colorectal cancer cells in vitro by inhibiting the PI3K signaling pathway. Sci Rep 2025; 15:15710. [PMID: 40325115 PMCID: PMC12052966 DOI: 10.1038/s41598-025-00480-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/28/2025] [Indexed: 05/07/2025] Open
Abstract
The occurrence and death rates of colorectal cancer (CRC) have been consistently increasing, demanding the immediate requirement for the creation of dependable and effective medicinal treatments. This study investigates bardoxolone methyl (CDDO-Me), a synthetic triterpenoid, as a potential therapeutic agent for CRC. Integrating network pharmacology, bioinformatic analyses, and in vitro assays, we evaluated the effects of CDDO-Me on CRC cells. In vitro studies indicate that CDDO-Me significantly inhibits the growth of CRC cells, specifically HCT116 and RKO, with IC50 values of 3.17 µM and 7.64 µM, respectively. Additionally, CDDO-Me effectively suppresses the PI3K signaling pathway and increases reactive oxygen species (ROS) levels in CRC cells. Consequently, this leads to G2/M phase arrest, an increase in apoptosis (over 30%), and a decrease in adhesion and invasion capabilities. Importantly, these effects were reversible upon the administration of N-acetylcysteine. Collectively, our findings provide compelling evidence for the potential of CDDO-Me as a promising candidate for the treatment of CRC.
Collapse
Affiliation(s)
- Linlin Zhang
- Department of Pharmacy, The First people's Hospital of Yancheng, Yancheng, 224000, China
| | - Qiaoling Huang
- The Third People's Hospital of Yueqing City Medical Community Beibaixiang Branch, Wenzhou, 325603, China
| | - Jingqing Yang
- Department of Pharmacy, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Ye Hu
- Department of Pharmacy, The First people's Hospital of Yancheng, Yancheng, 224000, China.
- Department of Pharmacy, Yancheng No.1 People's Hospital, Affiliated Hospital of Medical School, Nanjing University, Yancheng, 224000, China.
| |
Collapse
|
7
|
Gund V, Sharma S, Chandan S, Garg S, Singh S. Identification of non-synonymous SNPs affecting structure and function of MLH1 and NBN proteins: a computational approach. J Appl Genet 2025:10.1007/s13353-025-00968-2. [PMID: 40312597 DOI: 10.1007/s13353-025-00968-2] [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: 12/11/2024] [Revised: 04/10/2025] [Accepted: 04/12/2025] [Indexed: 05/03/2025]
Abstract
The genes NBN and MLH1 are critical for DNA repair, and this study aimed to detect and predict the effects of pathogenic single nucleotide polymorphisms (SNPs) in their mRNA and protein sequences. An in silico analysis assessed the impact of SNPs on the physicochemical properties, structure, stability, and function of MLH1 and NBN proteins. Results revealed that some SNPs significantly alter protein stability, structure, and binding interactions, potentially impairing DNA repair. Molecular docking studies further indicated disruptions in protein-protein interactions due to specific SNPs. These findings underscore the importance of using in silico methods to predict the functional effects of genetic variations, providing insights that could guide personalized treatments and improve cancer detection.
Collapse
Affiliation(s)
- Vaishnavi Gund
- Department of Biosciences and Bioengineering, D Y Patil International University Akurdi, Pune, 411044, Maharashtra, India
| | - Siddharth Sharma
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, Punjab, 147004, India
| | - Swet Chandan
- Department of Biosciences and Bioengineering, D Y Patil International University Akurdi, Pune, 411044, Maharashtra, India
| | - Shashank Garg
- Lovely Professional University, Phagwara, Punjab, India
| | - Sidhartha Singh
- Department of Biosciences and Bioengineering, D Y Patil International University Akurdi, Pune, 411044, Maharashtra, India.
| |
Collapse
|
8
|
Gong Y, Yuan X, Jiao Q, Yu Z. Unveiling fine-scale spatial structures and amplifying gene expression signals in ultra-large ST slices with HERGAST. Nat Commun 2025; 16:3977. [PMID: 40295488 PMCID: PMC12037780 DOI: 10.1038/s41467-025-59139-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
We propose HERGAST, a system for spatial structure identification and signal amplification in ultra-large-scale and ultra-high-resolution spatial transcriptomics data. To handle ultra-large spatial transcriptomics (ST) data, we consider the divide and conquer strategy and devise a Divide-Iterate-Conquer framework especially for spatial transcriptomics data analysis, which can also be adopted by other computational methods for extending to ultra-large-scale ST data analysis. To tackle the potential over-smoothing problem arising from data splitting, we construct a heterogeneous graph network to incorporate both local and global spatial relationships. In simulations, HERGAST consistently outperforms other methods across all settings with more than a 10% increase in average adjusted rand index (ARI). In real-world datasets, HERGAST's high-precision spatial clustering identifies SPP1+ macrophages intermingled within colorectal tumors, while the enhanced gene expression signals reveal unique spatial expression patterns of key genes in breast cancer.
Collapse
Affiliation(s)
- Yuqiao Gong
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Yuan
- SJTU-Yale Joint Center for Biostatistics and Data Science Organization, Shanghai Jiao Tong University, Shanghai, China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiong Jiao
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhangsheng Yu
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
- SJTU-Yale Joint Center for Biostatistics and Data Science Organization, Shanghai Jiao Tong University, Shanghai, China.
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Yang W, Cheng R, Qin M, Pan X, Tan Y, Feng K, Zhang J, Huang J. Tricellulin facilitates colorectal cancer metastasis through activation of the TGFβ/SMAD2/3 signalling pathway. Front Oncol 2025; 15:1562976. [PMID: 40291908 PMCID: PMC12021625 DOI: 10.3389/fonc.2025.1562976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
Background Tricellulin belongs to the TAMP family of proteins and is primarily localized at the tricellular tight junctions. While its role in the progression of cancer has been reported, its importance in the progression of colorectal cancer (CRC) remains unclear. Objective This study aimed to determine the function and mechanism of tricellulin in CRC progression. Methods The proteins expression in cells and/or tissues was determined by Western blot, immunohistochemistry staining, and/or RT-qPCR analyses. The biological functions of tricellulin were investigated through in vitro assays (CCK-8, Transwell migration, and colony formation assays) and in vivo xenograft models. Tricellulin was significantly upregulated in CRC tissues compared to adjacent normal tissues. The expression of tricellulin was correlated with poor prognosis in patients with CRC. Results In vitro assays showed that tricellulin enhanced CRC cell proliferation, migration, and invasion. Mechanistically, tricellulin activated the TGFb1/SMAD2/3 pathway, while TGFb1 reciprocally controlled the expression of tricellulin. Also, tricellulin promotes CRC cell migration/invasion through EMT. In vivo models confirmed that the overexpression of tricellulin facilitated tumor growth and activated the TGFb1/ SMAD2/3 pathway in CRC. Conclusion Our findings demonstrate thatTricellulin promotes the metastasis of colorectal cancer by activating the TGF-β/SMAD2/3 signaling pathway, and TGF-β1 can reciprocally regulate the expression of tricellulin.We have revealed a novel mechanism by which tricellulin forms a positive feedback loop to promote the growth and metastasis of CRC. This mechanism provides novel insights into CRC progression and suggests potential therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jinxiu Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jiean Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
10
|
Wang X, Lai J, Wang D, Wei K, Yang J. Immunophenotyping of colon cancer for identification of potential antigens for colon cancer vaccines. Front Oncol 2025; 15:1403256. [PMID: 40260289 PMCID: PMC12009704 DOI: 10.3389/fonc.2025.1403256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
Background Colon cancer is a prevalent malignancy that significantly threatens human health. In recent years, mRNA cancer vaccines have demonstrated considerable potential and distinct advantages in colon cancer treatment. Thus, This study identifies CUL7, ENO2, and MPP2 as potential antigens for colon cancer mRNA vaccines. Through multi-omics analysis, we classify COAD into three immune subtypes (C1-C3) with distinct molecular and clinical features. Methods Data from TCGA and GEO databases were analyzed using bioinformatics tools. Prognostic indices were calculated with GEPIA2, and TIMER assessed antigen-presenting cell infiltration. Survival analysis was performed using Kaplan-Meier curves and Cox proportional hazards models. Immune subtypes were classified via non-negative matrix factorization (NMF) clustering, with k=3 determined by cophenetic correlation (0.92) and silhouette width (average = 0.85). Drug sensitivity, immune cell infiltration, and gene set variation were analyzed using R packages such as "pRRophetic," CIBERSORT, and GSVA. Functional enrichment analysis was performed with GO, KEGG, and GSEA. Experimental validation included immunohistochemistry and RT-PCR to confirm gene expression. Results Analysis of TCGA-COAD data revealed copy number variants in 16,354 genes, with CUL7, ENO2, and MPP2 showing significant antigen-presenting cell infiltration and associations with overall survival (OS) and relapse-free survival (RFS). Based on molecular mechanisms, cellular features, and clinical characteristics, colon cancer was categorized into three immune subtypes (C1, C2, and C3) distinct from Thorsson's pan-cancer subtypes (C1-C6) in pathway enrichment, with the C2 subtype exhibited significantly longer overall survival (OS) than C1 and C3 (median OS: C2 = 68 months vs. C1 = 42 months, C3 = 37 months; log-rank P < 0.001). The distribution of these immune subtypes showed disparities in immune patterns, and a correlation between key components and immune cells was observed. Prognostic correlation analysis indicated that the gray and turquoise modules were closely linked to colorectal cancer prognosis. Additionally, RT-PCR confirmed the association of CUL7, ENO2, and MPP2 expression levels with colon cancer. Conclusions CUL7, ENO2, and MPP2 were identified as potential antigens for colon cancer mRNA vaccines, with MPP2 showing particular immunological relevance. This study provides a foundation for mRNA vaccine development and patient stratification for vaccination in colon cancer.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Oncology, The Third Affiliated Hospital of Shandong First Medical University (Affiliated Hospital of Shandong Academy of Medical Sciences), Jinan, Shandong, China
- The Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jingjiang Lai
- The Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Dawei Wang
- The Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Keyi Wei
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Jing Yang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| |
Collapse
|
11
|
Markov N, Sabirova S, Sharapova G, Gomzikova M, Brichkina A, Barlev NA, Egger M, Rizvanov A, Simon HU. Mitochondrial, metabolic and bioenergetic adaptations drive plasticity of colorectal cancer cells and shape their chemosensitivity. Cell Death Dis 2025; 16:253. [PMID: 40185729 PMCID: PMC11971274 DOI: 10.1038/s41419-025-07596-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
The extent of mitochondrial heterogeneity and the presence of mitochondrial archetypes in cancer remain unknown. Mitochondria play a central role in the metabolic reprogramming that occurs in cancer cells. This process adjusts the activity of metabolic pathways to support growth, proliferation, and survival of cancer cells. Using a panel of colorectal cancer (CRC) cell lines, we revealed extensive differences in their mitochondrial composition, suggesting functional specialisation of these organelles. We differentiated bioenergetic and mitochondrial phenotypes, which point to different strategies used by CRC cells to maintain their sustainability. Moreover, the efficacy of various treatments targeting metabolic pathways was dependent on the respiration and glycolysis levels of cancer cells. Furthermore, we identified metabolites associated with both bioenergetic profiles and cell responses to treatments. The levels of these molecules can be used to predict the therapeutic efficacy of anti-cancer drugs and identify metabolic vulnerabilities of CRC. Our study indicates that the efficacy of CRC therapies is closely linked to mitochondrial status and cellular bioenergetics.
Collapse
Affiliation(s)
- Nikita Markov
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Sirina Sabirova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Laboratory of Intercellular Communication, Kazan Federal University, Kazan, Russia
| | - Gulnaz Sharapova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Marina Gomzikova
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Laboratory of Intercellular Communication, Kazan Federal University, Kazan, Russia
| | - Anna Brichkina
- Institute of Systems Immunology, Center for Tumor Biology and Immunology, Philipps University of Marburg, Marburg, Germany
| | - Nick A Barlev
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Institute of Cytology of the Russian Academy of Sciences, St. Petersburg, Russia
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan
| | - Marcel Egger
- Department of Physiology, University of Bern, Bern, Switzerland
| | - Albert Rizvanov
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Kazan, Russia
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland.
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany.
| |
Collapse
|
12
|
Kang B, Qiao Y, Zhu J, Li J. Neoadjuvant therapy for colorectal cancer from 2015 to 2024: a visual analysis and bibliometric analysis. Front Oncol 2025; 15:1526610. [PMID: 40242245 PMCID: PMC11999843 DOI: 10.3389/fonc.2025.1526610] [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: 11/12/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Background Colorectal cancer (CRC) imposes a substantial burden on global health., but research trends and hotspots in this field are still not clear. The purpose of this research is to create a visual knowledge map based on bibliometric analysis, identify research hotspots and predict future research trends. Method Utilizing the Web of Science Core Collection (WoSCC) as data source and integrating the visualization capabilities of the Bibliometrix R software package, CiteSpace, and VOSviewer, analyze the authors, research institutions, countries, cited documents, publishing journals, abstracts, and keyword information of literature pertaining to neoadjuvant therapy for colorectal cancer spanning from January 2015 to December 2024. Result The analysis included 1,587 articles from 1,464 institutions, 385 journals, and 61 countries or regions. China has the largest number of publications (449) and the largest number of citations (5,035). The United States occupies the leading position with an average of 21.6. "Annals of Surgical Oncology" is the most published journal with 51 articles, and "Journal of Clinical Oncology" is the journal with the most references (4,465 references). Highly cited references focus on clinical trials and guidelines for neoadjuvant therapy for colorectal cancer. In recent years, the most important keywords in the research on colorectal cancer and neoadjuvant therapy have been "artificial intelligence", "total neoadjuvant therapy" and "immunotherapy". Conclusion This article provided a review of the research on neoadjuvant therapy for colorectal cancer, can provide reference for subsequent research on neoadjuvant therapy for colorectal cancer. The results offered valuable insights and data that informed the direction of future advancements.
Collapse
Affiliation(s)
- Boyu Kang
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
| | - Yihuan Qiao
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
| | - Jun Zhu
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
- Department of General Surgery, The Southern Theater Air Force Hospital, Guangzhou, China
| | - Jipeng Li
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi’an, China
- Department of Experiment Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
13
|
Yang JL, Ma JJ, Qu TY, Dai Q, Leng J, Fang L, Wu J, Li YJ, Yu HF. Glycolysis-related lncRNA FTX upregulates YAP1 to facilitate colorectal cancer progression via sponging miR-215-3p. Sci Rep 2025; 15:9929. [PMID: 40121300 PMCID: PMC11929783 DOI: 10.1038/s41598-025-94638-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
Increased evidence reveals that glycolysis is one of the key metabolic hallmarks of cancer cells. However, the roles of lncRNA FTX in energy metabolism and cancer progression remain unclear. In this study we aim to show that lncRNA FTX was significantly upregulated in cancer tissues and serum of CRC patients and CRC cell lines. Function study indicated that it could promote aerobic glycolysis, cell proliferation, migration and invasion in colorectal cancer cells. Further mechanistic studies showed, lncRNA FTX was found to function as a sponge for miR-215-3p, which reduced the ability of miR-215-3p to repress the YAP1 oncoprotein. Additionally, a negative correlation was observed between lncRNA FTX and miR-215-3p expression, and the knockdown of lncRNA FTX or miR-215-3p overexpression yielded opposite effects. In conclusion, this study demonstrates that FTX could directly combine with miR-215-3p as a competitive endogenous RNA, thus promoting the aerobic glycolysis and progression of CRC in vitro and in vivo.
Collapse
Affiliation(s)
- Jin-Lan Yang
- Department of Oncology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China
- Cancer Disease Research Institute, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 98# Fenghuang Road, Zunyi, 563003, Guizhou Province, China
| | - Jing-Jing Ma
- Department of Clinical Laboratory, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China
| | - Tian-Yin Qu
- Department of Oncology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China
- Cancer Disease Research Institute, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 98# Fenghuang Road, Zunyi, 563003, Guizhou Province, China
| | - Qing Dai
- Department of Oncology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China
- Cancer Disease Research Institute, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 98# Fenghuang Road, Zunyi, 563003, Guizhou Province, China
| | - Jing Leng
- Department of Oncology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China
- Cancer Disease Research Institute, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 98# Fenghuang Road, Zunyi, 563003, Guizhou Province, China
| | - Lin Fang
- Department of Oncology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China
- Cancer Disease Research Institute, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 98# Fenghuang Road, Zunyi, 563003, Guizhou Province, China
| | - Jie Wu
- Scientific Research Center, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou Province, China
| | - Ya-Jun Li
- Department of Oncology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China
| | - Huang-Fei Yu
- Department of Oncology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, Guizhou Province, China.
- Cancer Disease Research Institute, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 98# Fenghuang Road, Zunyi, 563003, Guizhou Province, China.
| |
Collapse
|
14
|
Wang T, Chen Z, Wang W, Wang H, Li S. Single-cell and spatial transcriptomic analysis reveals tumor cell heterogeneity and underlying molecular program in colorectal cancer. Front Immunol 2025; 16:1556386. [PMID: 40145096 PMCID: PMC11936967 DOI: 10.3389/fimmu.2025.1556386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Background Colorectal cancer (CRC) is a highly heterogeneous tumor, with significant variation in malignant cells, posing challenges for treatment and prognosis. However, this heterogeneity offers opportunities for personalized therapy. Methods The consensus non-negative matrix factorization algorithm was employed to analyze single-cell transcriptomic data from CRC, which helped identify malignant cell expression programs (MCEPs). Subsequently, a crosstalk network linking MCEPs with immune/stromal cell trajectory development was constructed using Monocle3 and NicheNet. Additionally, bulk RNA-seq data were utilized to systematically explore the relationships between MCEPs, clinical features, and genetic mutations. A prognostic model was then established through Lasso and Cox regression analyses, integrating clinical data into a nomogram for personalized risk prediction. Furthermore, key genes associated with MCEPs and their potential therapeutic targets were identified using protein-protein interaction networks, followed by molecular docking to predict drug-binding affinity. Results We classified CRC malignant cell transcriptional states into eight distinct MCEPs and successfully constructed crosstalk networks between these MCEPs and immune or stromal cells. A prognostic model containing 15 genes was developed, demonstrating an AUC greater than 0.8 for prognostic evaluation over 1 to 10 years when combined with clinical features. A key drug target gene TIMP1 was identified, and several potential targeted drugs were discovered. Conclusion This study demonstrated that characterization of the malignant cell transcriptional programs could effectively reveal the biological features of highly heterogeneous tumors like CRC and exhibit significant potential in tumor prognosis assessment. Our research provides new theoretical and practical directions for CRC prognosis and targeted therapy.
Collapse
Affiliation(s)
- Teng Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Zhaoming Chen
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Wang Wang
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, China
| | - Heng Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Shenglong Li
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
Sun J, Chen Y, Xu Z, Wang W, Li P. Notch signaling in the tumor immune microenvironment of colorectal cancer: mechanisms and therapeutic opportunities. J Transl Med 2025; 23:315. [PMID: 40075484 PMCID: PMC11900264 DOI: 10.1186/s12967-025-06282-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related morbidity and mortality worldwide, driven by a complex interplay of genetic, environmental, and immune-related factors. Among the pivotal pathways implicated in CRC tumorigenesis, the Notch signaling pathway is instrumental in governing cell fate decisions, tissue renewal, homeostasis, and immune cell development. As a highly conserved mechanism, Notch signaling not only modulates tumor cell behavior but also shapes the immune landscape within the tumor microenvironment (TME). Aberrant Notch signaling in CRC fosters immune evasion and tumor progression through its effects on the balance and functionality of immune cells, including myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs). Elevated Notch pathway activation correlates with advanced clinicopathological features and poorer clinical outcomes, highlighting its relevance as both a prognostic biomarker and a therapeutic target. Therapeutic approaches aimed at inhibiting the Notch pathway, such as γ-secretase inhibitors (GSIs) or monoclonal antibodies (mAbs) in combination with other therapies, have demonstrated promising efficacy in preclinical and clinical settings. This review examines the impact of Notch signaling on CRC immunity, elucidating its regulatory mechanisms within immune cells and its role in promoting tumor progression. Additionally, this review discusses therapeutic strategies targeting Notch signaling, including GSIs, mAbs, and potential combination therapies designed to overcome resistance and improve patient outcomes. By elucidating the multifaceted role of Notch within the CRC TME, this review underscores its potential as a target for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Jiachun Sun
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, 471003, China
| | - Yi Chen
- Department of Children's gastroenterology, Anhui Children's Hospital, Hefei, Anhui, 230000, China
| | - Ziyi Xu
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, 471003, China
| | - Weizheng Wang
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, 471003, China
| | - Penghui Li
- Department of Gastrointestinal surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, No. 24 Jinghua Road, Jianxi District, Luoyang, Henan, 471000, China.
| |
Collapse
|
16
|
Kogai H, Tsukamoto S, Koga M, Miyano M, Akagi T, Yamaguchi A, Mori K, Gotoh K, Nakazawa Y. Broad-Spectrum Efficacy of CEACAM6-Targeted Antibody-Drug Conjugate with BET Protein Degrader in Colorectal, Lung, and Breast Cancer Mouse Models. Mol Cancer Ther 2025; 24:392-405. [PMID: 39812376 PMCID: PMC11876960 DOI: 10.1158/1535-7163.mct-24-0444] [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: 06/08/2024] [Revised: 10/24/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Despite remarkable advances in cancer treatment, most solid cancers remain difficult to cure. We recently developed an antibody-drug conjugate (ADC; 84-EBET) for pancreatic cancer by using the carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) antibody #84.7 and the bromodomain and extra-terminal (BET) protein degrader EBET. In this study, we showed the overexpression of CEACAM6 in colorectal, lung, and breast cancers and the broad-spectrum efficacy of 84-EBET in mouse models of these cancers. In vitro assays using cancer organoids and cell lines of colorectal, lung, and breast cancers revealed that 84-EBET was more potent than ADCs with known approved payloads-DXd, SN38, and monomethyl auristatin E-or standard chemotherapies. In mouse studies, a single injection of 84-EBET induced marked regression of colorectal-, lung-, and breast cancer patient-derived xenograft tumors and cell line-derived xenograft tumors. Moreover, in mouse syngeneic colorectal cancer, lung cancer, and breast cancer models resistant to PD-1 antibody, the combination of 84-EBET and PD-1 antibody induced complete regression of most tumors. Mechanistically, 84-EBET degraded bromodomain-containing protein 4 in both cancer and stromal cells via bystander efficacy. It decreased stromal inflammatory phenotypes and increased activated T-cell numbers in tumors. These results demonstrate that delivering BET protein degraders to tumors and their microenvironments via a CEACAM6-targeted ADC may be effective against a wide range of solid cancers.
Collapse
Affiliation(s)
- Hiroyuki Kogai
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | | | - Minaho Koga
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | - Masayuki Miyano
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | - Tsuyoshi Akagi
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | | | - Kiyoshi Mori
- Department of Central Laboratory and Surgical Pathology, NHO Osaka National Hospital, Osaka, Japan
| | - Kunihito Gotoh
- Department of Surgery, NHO Osaka National Hospital, Osaka, Japan
| | - Youya Nakazawa
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| |
Collapse
|
17
|
Xu Y, Zhou J, Wu Y, Shen J, Fu X, Liu M, Liang S. New insights into the role of complement system in colorectal cancer (Review). Mol Med Rep 2025; 31:68. [PMID: 39791217 PMCID: PMC11751662 DOI: 10.3892/mmr.2025.13433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide. With the growing understanding of immune regulation in tumors, the complement system has been recognized as a key regulator of tumor immunity. Traditionally, the complement cascade, considered an evolutionarily conserved defense mechanism against invading pathogens, has been viewed as a crucial inhibitor of tumor progression. Complement components or activation products produced via cascade‑dependent or ‑independent processes are associated with the regulation of tumor‑associated inflammation. Various forms of complement activation products present in body fluids or inside cells, along with complement regulatory proteins and complement receptors, are involved in tumor cell growth and modulating the tumor microenvironment. In the present review, the role of the complement system in the tumor immunity of CRC is discussed. In addition, the contribution of the unconventional cascade‑independent pathway of complement activation in CRC progression is highlighted. A deeper understanding of the mechanism underlying the complement system in colitis‑associated colorectal cancer (CAC) may provide novel insights to assist the development of methods to prevent tumor progression and identify potential targets for the treatment of CAC.
Collapse
Affiliation(s)
- Yuwen Xu
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Jiaqi Zhou
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Yuanyuan Wu
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Jie Shen
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Xiaoyan Fu
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Meifang Liu
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Shujuan Liang
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
18
|
Ying L, Zhang L, Chen Y, Huang C, Zhou J, Xie J, Liu L. Predicting immunotherapy prognosis and targeted therapy sensitivity of colon cancer based on a CAF-related molecular signature. Sci Rep 2025; 15:6387. [PMID: 39984646 PMCID: PMC11845748 DOI: 10.1038/s41598-025-90899-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/17/2025] [Indexed: 02/23/2025] Open
Abstract
The role of cancer-associated fibroblasts (CAFs) in modulating the tumor microenvironment (TME) is gaining attention, yet their impact on prognosis and therapeutic response in colon cancer remains unclear. Here, we identified genes associated with CAF infiltration via weighted gene co-expression network analysis (WGCNA) utilizing data from The Cancer Genome Atlas (TCGA) and GSE39582 cohorts. Univariate Cox regression and least absolute shrinkage and selection operator (LASSO) regression analyses were used to construct CAF molecular signatures (CAFscore). Patients were categorized into high and low CAFscore groups to analyze clinicopathological traits, somatic mutations, immune evasion, and treatment responses. In this study, a total of 244 genes were correlated with CAF infiltration, with 11 linked to overall survival. Notably, FSTL3, CRIP2, and SLC2A3 were selected for the CAFscore. A higher CAFscore was associated with poorer prognoses, increased malignancy, and therapeutic resistance, particularly among patients with high tumor mutation burden and microsatellite instability. Furthermore, elevated FSTL3 expression was associated with reduced CD8+ T cell infiltration, indicating a suppressive TME. Mechanistically, CAFs may promote immune evasion via NAMPT ligand-receptor interactions based on single-cell RNA sequencing data. Thus, the CAFscore is crucial for personalizing treatment strategies and identifying patients who require more aggressive management.
Collapse
Affiliation(s)
- Leqian Ying
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
- School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Lu Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
- School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Yanping Chen
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Chunchun Huang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
- School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Jingyi Zhou
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
- School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Jinbing Xie
- Department of Radiology, Nurturing Center of Jiangsu Province for the State Laboratory of AI Imaging and Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China
| | - Lin Liu
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao, Nanjing, 210000, China.
| |
Collapse
|
19
|
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
|
20
|
Sun L, Huangfu L, Li F, Yan Y, Kong R, Ji K, Li J. Identification and functional characterization of m1A-related genes in colorectal cancer: implications for prognosis, immune infiltration, and therapeutic strategies. Front Oncol 2025; 15:1532602. [PMID: 40040720 PMCID: PMC11876182 DOI: 10.3389/fonc.2025.1532602] [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: 11/22/2024] [Accepted: 01/24/2025] [Indexed: 03/06/2025] Open
Abstract
Introduction Colorectal cancer (CRC), characterized by its complex genetic heterogeneity and varied responses to treatment, is a leading cause of cancer-related mortality worldwide. The role of N1-methyladenosine (m1A)-related genes in tumor biology remains underexplored. This study aimed to investigate the prognostic value of m1A-related genes in CRC, characterize their role in tumor molecular subtyping, and explore their influence on the tumor microenvironment (TME) and immune infiltration. Methods To identify prognostic markers, univariate Cox analysis was performed using multiple datasets, including TCGA and GEO, identifying 43 m1A-related genes. Four distinct molecular subtypes of CRC were defined based on the expression of these genes using non-negative matrix factorization (NMF). Immune infiltration analysis was conducted, and the TIDE algorithm was used to predict response to immune checkpoint inhibitors (ICIs). Furthermore, a prognostic model based on m1A-related genes was constructed and validated across multiple datasets. Results The results demonstrated that the four CRC molecular subtypes exhibited significant differences in survival outcomes and clinical characteristics. Stromal cells showed higher m1A scores, suggesting a regulatory role in the TME. There was a positive correlation between m1A-related gene expression and immune checkpoint genes. Moreover, the constructed prognostic model showed robust predictive performance and outperformed other recently published models. Discussion The findings suggest that m1A-related genes are not only valuable biomarkers for CRC prognosis but also have significant implications for the immune landscape and could serve as potential targets for therapeutic intervention, particularly in the context of immunotherapy. For instance, SLC12A2 was found to enhance invasion, proliferation, and migration of colorectal cancer cells while inhibiting apoptosis. Further studies are needed to understand the functional roles of m1A modifications across different cell types within the TME.
Collapse
Affiliation(s)
- Lan Sun
- Department of Pharmaceutical Engineering, Jiangsu Food and Pharmaceutical Science College, Huaian, Jiangsu, China
| | - Liwei Huangfu
- Department of Pharmaceutical Engineering, Jiangsu Food and Pharmaceutical Science College, Huaian, Jiangsu, China
| | - Fang Li
- State Key Laboratory of New-tech For Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu, China
| | - Yuhui Yan
- Department of Pharmaceutical Engineering, Jiangsu Food and Pharmaceutical Science College, Huaian, Jiangsu, China
| | - Ruiping Kong
- Department of Pharmaceutical Engineering, Jiangsu Food and Pharmaceutical Science College, Huaian, Jiangsu, China
| | - Kun Ji
- Department of Pharmaceutical Engineering, Jiangsu Food and Pharmaceutical Science College, Huaian, Jiangsu, China
| | - Jiachun Li
- Department of Grain and Food Pharmacy, Jiangsu Vocational College of Finance and Economics, Huaian, Jiangsu, China
| |
Collapse
|
21
|
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
|
22
|
Li KP, Wan S, Wang CY, Chen SY, Wang L, Liu SH, Yang L. Multi-omics analysis reveals the impact of YAP/TEAD4-mediated EIF5A1 expression on mitochondrial apoptosis and bladder cancer progression. BMC Cancer 2025; 25:234. [PMID: 39934701 PMCID: PMC11817321 DOI: 10.1186/s12885-025-13522-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 01/14/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Eukaryotic Initiation Factor 5A1 (EIF5A1) is a translation factor, and its pro-tumorigenic role has been extensively documented across various cancer types. However, its specific function in bladder cancer (BLCA) remains unclear. METHODS We integrated proteomics and transcriptomics data with clinical data from BLCA patients to investigate the correlation between EIF5A1 expression and BLCA, as well as its potential clinical applications. Transcriptomic data were employed to explore the downstream signaling pathways regulated by EIF5A1. Furthermore, ChIP analysis and luciferase reporter assays were conducted to identify the upstream transcription factors regulating EIF5A1. RESULTS EIF5A1 expression is significantly upregulated in cancer tissues and cells and is strongly associated with poor prognosis. Silencing EIF5A1 in BLCA cells significantly reduced invasiveness, and proliferative capacity. Mechanistic studies identified YAP/TEAD4 as a transcription factor that regulates EIF5A1, influencing mitochondrial-mediated apoptosis by activating the JAK2/STAT3 signaling pathway, thereby promoting BLCA progression. CONCLUSION Our research demonstrates that EIF5A1 is upregulated in BLCA and associated with poor prognosis. We identified TEAD4 as a potential transcriptional regulator of EIF5A1 and showed that EIF5A1 expression is associated with changes in JAK2/STAT3 signaling and mitochondrial apoptosis in BLCA.
Collapse
Affiliation(s)
- Kun-Peng Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, Lanzhou, China
| | - Shun Wan
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, Lanzhou, China
| | - Chen-Yang Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, Lanzhou, China
| | - Si-Yu Chen
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, Lanzhou, China
| | - Li Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, Lanzhou, China
| | - Shan-Hui Liu
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China.
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, Lanzhou, China.
| | - Li Yang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China.
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, Lanzhou, China.
| |
Collapse
|
23
|
Puzzo M, De Santo M, Morelli C, Leggio A, Catalano S, Pasqua L. Colorectal Cancer: Current and Future Therapeutic Approaches and Related Technologies Addressing Multidrug Strategies Against Multiple Level Resistance Mechanisms. Int J Mol Sci 2025; 26:1313. [PMID: 39941081 PMCID: PMC11818749 DOI: 10.3390/ijms26031313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/31/2025] [Accepted: 02/01/2025] [Indexed: 02/16/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and is associated with a poor prognosis. The mutation profile and related involved pathways of CRC have been, in broad terms, analyzed. The main current therapeutic approaches have been comprehensively reviewed here, and future possible therapeu-tic options and related technologies have been perspectively presented. The complex scenario represented by the multiple-level resistance mechanism in the epidermal growth factor receptor (EGFR) pathway, including mutations in KRAS, NRAS, and BRAF V600E, is discussed. Examples of engineered therapeutic approaches from the literature along with a drug combination tested in clinical trials are discussed. The encouraging results observed with the latter combination (the BEACON clinical trial), totally free from chemotherapy, prompted the authors to imagine a future possible nanotechnology-assisted therapeutic approach for bypassing multiple-level resistance mechanisms, hopefully allowing, in principle, a complete biological cancer remission.
Collapse
Affiliation(s)
- Marianna Puzzo
- Laboratory of Clinical, Biomolecular and Genetic Analyses Unit, Annunziata Hospital, 87100 Cosenza, Italy; (M.P.); (S.C.)
| | - Marzia De Santo
- Department of Pharmacy, Health and Nutritional Sciences University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy; (M.D.S.); (C.M.); (A.L.)
- NanoSiliCal Devices s.r.l., University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Catia Morelli
- Department of Pharmacy, Health and Nutritional Sciences University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy; (M.D.S.); (C.M.); (A.L.)
- NanoSiliCal Devices s.r.l., University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Antonella Leggio
- Department of Pharmacy, Health and Nutritional Sciences University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy; (M.D.S.); (C.M.); (A.L.)
- NanoSiliCal Devices s.r.l., University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Stefania Catalano
- Laboratory of Clinical, Biomolecular and Genetic Analyses Unit, Annunziata Hospital, 87100 Cosenza, Italy; (M.P.); (S.C.)
- Department of Pharmacy, Health and Nutritional Sciences University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy; (M.D.S.); (C.M.); (A.L.)
| | - Luigi Pasqua
- NanoSiliCal Devices s.r.l., University of Calabria, 87036 Arcavacata di Rende, Italy
- Department of Environmental Engineering, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy
| |
Collapse
|
24
|
Melino G, Bischof J, Chen WL, Jia W, Juhl H, Kopeina GS, Mauriello A, Novelli F, Scimeca M, Shi Y, Pirozzi BM, Sica G, Zamaraev AV, Zhivotovsky B. New hope for the world cancer day. Biol Direct 2025; 20:14. [PMID: 39901154 PMCID: PMC11792201 DOI: 10.1186/s13062-025-00608-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025] Open
Affiliation(s)
- Gerry Melino
- TOR, University of Rome Tor Vergata, Rome, 00133, Italy.
| | - Julia Bischof
- Indivumed GmbH, Falkenried, 88 Building D, 20251, Hamburg, Germany
| | - Wen-Lian Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Wei Jia
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China.
| | - Harmut Juhl
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China.
| | - Gelina S Kopeina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | | | - Flavia Novelli
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI, 96816, USA
| | | | - Yufang Shi
- The Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| | | | - Giuseppe Sica
- TOR, University of Rome Tor Vergata, Rome, 00133, Italy.
| | - Alexey V Zamaraev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Boris Zhivotovsky
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
- Lomonosov Moscow State University, Moscow, Russia.
- Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
25
|
Chen R, He A, De Carvalho DD. Viral mimicry evasion: a new role for oncogenic KRAS mutations. Mol Oncol 2025; 19:271-274. [PMID: 39592415 PMCID: PMC11792985 DOI: 10.1002/1878-0261.13771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
"Viral mimicry" refers to the induction of an innate immune response and interferon signaling by endogenous stimuli such as double-stranded RNA (dsRNA). This response has been shown to have strong cancer therapeutic potential, including by enhancing the effectiveness of immune checkpoint inhibition (ICI) therapies, and may represent a tumor suppression mechanism that needs to be overcome for malignant transformation to proceed. In a recent study, Zhou and colleagues identify KRAS, a frequently mutated oncogene, as a negative regulator of dsRNA and viral mimicry in an ICI-resistant colorectal cancer model. Oncogenic KRASG12D mutations downregulate the RNA-binding protein DDX60 by activating the AKT signaling pathway, which inhibits STAT3, a critical transcription factor regulating DDX60 and other interferon-stimulated genes. Overexpression of DDX60, which competitively binds to dsRNA to prevent RISC-mediated degradation, or targeting of KRASG12D elevated dsRNA levels, resulting in viral mimicry activation and potentiation of ICI treatment. These results establish KRAS as a promising target to sensitize immune "cold" tumors to ICI therapy and demonstrate the potential role of oncogenic mutations in viral mimicry evasion during tumorigenesis.
Collapse
Affiliation(s)
- Raymond Chen
- Department of Medical BiophysicsUniversity of TorontoCanada
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoCanada
| | - Aobo He
- Department of Medical BiophysicsUniversity of TorontoCanada
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoCanada
| | - Daniel D. De Carvalho
- Department of Medical BiophysicsUniversity of TorontoCanada
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoCanada
| |
Collapse
|
26
|
Vlachavas EI, Voutetakis K, Kosmidou V, Tsikalakis S, Roditis S, Pateas K, Kim R, Pagel K, Wolf S, Warsow G, Dimitrakopoulou-Strauss A, Zografos GN, Pintzas A, Betge J, Papadodima O, Wiemann S. Molecular and functional profiling unravels targetable vulnerabilities in colorectal cancer. Mol Oncol 2025. [PMID: 39876058 DOI: 10.1002/1878-0261.13814] [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: 06/26/2024] [Revised: 11/11/2024] [Accepted: 01/20/2025] [Indexed: 01/30/2025] Open
Abstract
Colorectal cancer (CRC) patients with microsatellite-stable (MSS) tumors are mostly treated with chemotherapy. Clinical benefits of targeted therapies depend on mutational states and tumor location. Many tumors carry mutations in KRAS proto-oncogene, GTPase (KRAS) or B-Raf proto-oncogene, serine/threonine kinase (BRAF), rendering them more resistant to therapies. We performed whole-exome sequencing and RNA-Sequencing of 28 tumors of the Athens Comprehensive Cancer Center CRC cohort, and molecularly characterized CRC patients based on their microsatellite instability (MSI) status, single-nucleotide variations (SNVs)/copy number alterations (CNAs), and pathway/transcription factor activities at the individual patient level. Variants were classified using a computational score for integrative cancer variant annotation and prioritization. Complementing this with public multi-omics datasets, we identified activation of transforming growth factor beta (TGFβ) signaling to be more strongly activated in MSS patients, whereas Janus kinase (JAK)-signal transducer and activator of transcription (STAT) and mitogen-activated protein kinase (MAPK) molecular cascades were activated specifically in MSI tumors. We unraveled mechanisms consistently perturbed in the transcriptional and mutational circuits and identified Runt-related transcription factors (RUNX transcription factors) as putative biomarkers in CRC, given their role in the regulation of pathways involved in tumor progression and immune evasion. Assessing the immunogenicity of CRC tumors in the context of RAS/RAF mutations and MSI/MSS status revealed a critical impact that KRAS mutations have on immunogenicity, particularly in the MSS patient subgroup, with implications for diagnosis and treatment.
Collapse
Affiliation(s)
| | | | - Vivian Kosmidou
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Spyridon Tsikalakis
- Division of Molecular Genome Analysis, German Cancer Research Center, Heidelberg, Germany
| | - Spyridon Roditis
- 3rd Surgical Department G.Gennimatas Hospital, Athens, Greece
- Surgical Department, University Hospital of North Midlands, Stoke-on-Trent, UK
| | | | | | | | - Stephan Wolf
- High-Throughput Sequencing Core Facility, German Cancer Research Center, Heidelberg, Germany
| | - Gregor Warsow
- Omics IT and Data Management Core Facility, German Cancer Research Center, Heidelberg, Germany
| | | | | | - Alexander Pintzas
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Johannes Betge
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Germany
- DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Germany
| | - Olga Papadodima
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
27
|
Colombo A, Concetta PM, Gebbia V, Sambataro D, Scandurra G, Valerio MR. A Narrative Review of the Role of Immunotherapy in Metastatic Carcinoma of the Colon Harboring a BRAF Mutation. In Vivo 2025; 39:25-36. [PMID: 39740863 PMCID: PMC11705148 DOI: 10.21873/invivo.13802] [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: 09/04/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 01/02/2025]
Abstract
Patients affected by metastatic carcinoma of the colon/rectum (mCRC) harboring mutations in the BRAF gene (MBRAF) respond poorly to conventional therapy and have a prognosis worse than that of patients without mutations. Despite the promising outcomes of targeted therapy utilizing multi-targeted inhibition of the mitogen-activated protein kinase (MAPK) signaling system, the therapeutic efficacy, especially for the microsatellite stable/DNA proficient mismatch repair (MSS/PMMR) subtype, remains inadequate. Patients with MBRAF/mCRC and high microsatellite instability or DNA deficient mismatch repair (MSI-H/DMMR) exhibit a substantial tumor mutation burden, suggesting a high probability of response to immunotherapy. It is widely acknowledged that MSS/pMMR/mCRC is an immunologically "cold" malignancy that exhibits resistance to immunotherapy. The integration of targeted therapy and immunotherapy may enhance clinical outcomes in patients with MBRAF/mCRC. Efforts to enhance outcomes are exclusively focused on MSS/DMMR-BRAF mutant cancers, which constitute the largest proportion. This review evaluates the clinical efficacy and advancement of novel immune checkpoint blockade therapies for MSI-H/DMMR and MSS/PMMR BRAF mutant mCRC. We examine potential indicators in the tumor immune milieu for forecasting immunotherapeutic response in BRAF mutant mCRC.
Collapse
Affiliation(s)
| | | | - Vittorio Gebbia
- Medical Oncology, Department of Medicine and Surgery, Kore University of Enna, Enna, Italy;
- Medical Oncology Unit, CdC Torina, Palermo, Italy
| | - Daniela Sambataro
- Medical Oncology, Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
- Medical Oncology Unit, Ospedale Umberto I, Enna, Italy
| | - Giuseppina Scandurra
- Medical Oncology, Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
- Medical Oncology Unit, Ospedale Cannizzario, Catania, Italy
| | | |
Collapse
|
28
|
Ferkel SAM, Holman EA, Sojwal RS, Rubin SJS, Rogalla S. Tumor-Infiltrating Immune Cells in Colorectal Cancer. Neoplasia 2025; 59:101091. [PMID: 39642846 DOI: 10.1016/j.neo.2024.101091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 12/09/2024]
Abstract
Colorectal cancer encompasses a heterogeneous group of malignancies that differ in pathophysiological mechanisms, immune response and infiltration, therapeutic response, and clinical prognosis. Numerous studies have highlighted the clinical relevance of tumor-infiltrating immune cells among different types of colorectal tumors yet vary in cell type definitions and cell identification strategies. The distinction of immune signatures is particularly challenging when several immune subtypes are involved but crucial to identify novel intercellular mechanisms within the tumor microenvironment. In this review, we compile human and non-human studies on tumor-infiltrating immune cells and provide an overview of immune subtypes, their pathophysiological functions, and their prognostic role in colorectal cancer. We discuss how differentiating immune signatures can guide the development of immunotherapeutic targets and personalized treatment regimens. We analyzed comprehensive human protein biomarker profiles across the entire immune spectrum to improve interpretability and application of tumor studies and to ultimately enhance immunotherapy and advance precision medicine for colorectal cancer patients.
Collapse
Affiliation(s)
- Sonia A M Ferkel
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Elizabeth A Holman
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Raoul S Sojwal
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Samuel J S Rubin
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA
| | - Stephan Rogalla
- Stanford University, School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, USA.
| |
Collapse
|
29
|
Li J, Wang Y, Xu J, Niu M, Wan S, Liu Y, Ding Z, Li G, Qian Q, Li D. The Expression Characteristics of the RBFOX1 Gene in Colorectal Cancer. Technol Cancer Res Treat 2025; 24:15330338251333695. [PMID: 40395202 DOI: 10.1177/15330338251333695] [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] [Indexed: 05/22/2025] Open
Abstract
IntroductionCopy number variation is a significant characteristic of colorectal cancer progression. RBFOX1 (A2BP1) is the gene with the highest frequency of copy number loss in colorectal cancer, but current research related to it and colorectal cancer is relatively scarce.MethodsData from TCGA and other sources were used to analyze the copy number variation and mRNA expression levels of RBFOX1, as well as their correlation with clinical pathological data. Immunohistochemistry and immunofluorescence experiments were used to analyze the expression of RBFOX1 protein in colorectal cancer cells and tissues.ResultsRBFOX1 has a high frequency (22.4%) of copy number loss and diverse copy number variations in colorectal cancer tissues. High-level RBFOX1 deletion is prone to occur in the right-sided colon and tissues with high microsatellite instability. The copy number variation of RBFOX1 and mRNA expression are not correlated. In tumor tissues, RBFOX1 mRNA shows a characteristic of reduced expression, which is significantly related to BRAF mutation (P = 4.7e-05, P = 0.03). Low expression of RBFOX1 is prone to occur in the right-sided colon and tissues with high microsatellite instability. The protein encoded by RBFOX1 is expressed in normal intestinal tissues, but shows a characteristic of absence in some colorectal cancer tissues.ConclusionIn the right-sided colon and tissues with high microsatellite instability, RBFOX1 shows copy number loss and low mRNA expression. This characteristic is closely related to BRAF gene mutation, and the protein of RBFOX1 is absent in some colorectal cancer tissues.
Collapse
Affiliation(s)
- Jian Li
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University
- Wuhan Clinical Research Center for Constipation and Pelvic Floor Disorders
- Quality Control Center of Colorectal and Anal Surgery of Health Commission of Hubei Province, Wuhan, Hubei Province, China
- JingMen People's Hospital, Jingchu University of Technology Affiliated JingMen People's Hospital
| | - Youheng Wang
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University
- Wuhan Clinical Research Center for Constipation and Pelvic Floor Disorders
- Quality Control Center of Colorectal and Anal Surgery of Health Commission of Hubei Province, Wuhan, Hubei Province, China
| | - Jian Xu
- Department of Pathology of Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Min Niu
- Operating Room of Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Songlin Wan
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University
- Wuhan Clinical Research Center for Constipation and Pelvic Floor Disorders
- Quality Control Center of Colorectal and Anal Surgery of Health Commission of Hubei Province, Wuhan, Hubei Province, China
| | - Yi Liu
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University
- Wuhan Clinical Research Center for Constipation and Pelvic Floor Disorders
- Quality Control Center of Colorectal and Anal Surgery of Health Commission of Hubei Province, Wuhan, Hubei Province, China
| | - Zhao Ding
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University
- Wuhan Clinical Research Center for Constipation and Pelvic Floor Disorders
- Quality Control Center of Colorectal and Anal Surgery of Health Commission of Hubei Province, Wuhan, Hubei Province, China
| | - Guangchun Li
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University
- Wuhan Clinical Research Center for Constipation and Pelvic Floor Disorders
- Quality Control Center of Colorectal and Anal Surgery of Health Commission of Hubei Province, Wuhan, Hubei Province, China
| | - Qun Qian
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University
- Wuhan Clinical Research Center for Constipation and Pelvic Floor Disorders
- Quality Control Center of Colorectal and Anal Surgery of Health Commission of Hubei Province, Wuhan, Hubei Province, China
| | - Daojiang Li
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University
- Wuhan Clinical Research Center for Constipation and Pelvic Floor Disorders
- Quality Control Center of Colorectal and Anal Surgery of Health Commission of Hubei Province, Wuhan, Hubei Province, China
| |
Collapse
|
30
|
Gong J, Lee C, Kim H, Kim J, Jeon J, Park S, Cho K. Control of Cellular Differentiation Trajectories for Cancer Reversion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2402132. [PMID: 39661721 PMCID: PMC11744559 DOI: 10.1002/advs.202402132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 11/08/2024] [Indexed: 12/13/2024]
Abstract
Cellular differentiation is controlled by intricate layers of gene regulation, involving the modulation of gene expression by various transcriptional regulators. Due to the complexity of gene regulation, identifying master regulators across the differentiation trajectory has been a longstanding challenge. To tackle this problem, a computational framework, single-cell Boolean network inference and control (BENEIN), is presented. Applying BENEIN to human large intestinal single-cell transcriptome data, MYB, HDAC2, and FOXA2 are identified as the master regulators whose inhibition induces enterocyte differentiation. It is found that simultaneous knockdown of these master regulators can revert colorectal cancer cells into normal-like enterocytes by synergistically inducing differentiation and suppressing malignancy, which is validated by in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Jeong‐Ryeol Gong
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Chun‐Kyung Lee
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Hoon‐Min Kim
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Juhee Kim
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Jaeog Jeon
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Sunmin Park
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Kwang‐Hyun Cho
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| |
Collapse
|
31
|
Sankaran S, Dubey R, Gomatam A, Chakor R, Kshirsagar A, Lohidasan S. Deciphering the multi-functional role of Indian propolis for the management of Alzheimer's disease by integrating LC-MS/MS, network pharmacology, molecular docking, and in-vitro studies. Mol Divers 2024; 28:4325-4342. [PMID: 38466554 DOI: 10.1007/s11030-024-10818-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/27/2024] [Indexed: 03/13/2024]
Abstract
The conventional one-drug-one-disease theory has lost its sheen in multigenic diseases such as Alzheimer's disease (AD). Propolis, a honeybee-derived product has ethnopharmacological evidence of antioxidant, anti-inflammatory, antimicrobial and neuroprotective properties. However, the chemical composition is complex and highly variable geographically. So, to leverage the potential of propolis as an effective treatment modality, it is essential to understand the role of each phytochemical in the AD pathophysiology. Therefore, the present study was aimed at investigating the anti-Alzheimer effect of bioactive in Indian propolis (IP) by combining LC-MS/MS fingerprinting, with network-based analysis and experimental validation. First, phytoconstituents in IP extract were identified using an in-house LC-MS/MS method. The drug likeness and toxicity were assessed, followed by identification of AD targets. The constituent-target-gene network was then constructed along with protein-protein interactions, gene pathway, ontology, and enrichment analysis. LC-MS/MS analysis identified 16 known metabolites with druggable properties except for luteolin-5-methyl ether. The network pharmacology-based analysis revealed that the hit propolis constituents were majorly flavonoids, whereas the main AD-associated targets were MAOB, ESR1, BACE1, AChE, CDK5, GSK3β, and PTGS2. A total of 18 gene pathways were identified to be associated, with the pathways related to AD among the topmost enriched. Molecular docking analysis against top AD targets resulted in suitable binding interactions at the active site of target proteins. Further, the protective role of IP in AD was confirmed with cell-line studies on PC-12, in situ AChE inhibition, and antioxidant assays.
Collapse
Affiliation(s)
- Sandeep Sankaran
- Department of Quality Assurance Techniques, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Rahul Dubey
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Anish Gomatam
- Department of Pharmaceutical Chemistry, Bombay College of Pharmacy, Mumbai, Maharashtra, 400098, India
| | - Rishikesh Chakor
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Ashwini Kshirsagar
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Sathiyanarayanan Lohidasan
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India.
| |
Collapse
|
32
|
Liu M, Liu Q, Hu K, Dong Y, Sun X, Zou Z, Ji D, Liu T, Yu Y. Colorectal cancer with BRAF V600E mutation: Trends in immune checkpoint inhibitor treatment. Crit Rev Oncol Hematol 2024; 204:104497. [PMID: 39245296 DOI: 10.1016/j.critrevonc.2024.104497] [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: 04/02/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/10/2024] Open
Abstract
Colorectal cancer (CRC) with BRAF V600E mutation presents a formidable scientific and clinical challenge due to its aggressive nature and poor response to standard therapeutic approaches. BRAF V600E mutation-induced conspicuous activation of the MAPK pathway contributes to the relentless tumor progression. Nevertheless, the efficacy of multi-targeted MAPK pathway inhibition remains suboptimal in clinical practice. Patients with high microsatellite instability (MSI-H) have shown favorable results with immune checkpoint inhibitors (ICIs). The combination of the MAPK pathway inhibition with ICIs has recently emerged as a promising regimen to improve clinical outcomes in the microsatellite stable (MSS) subgroup of BRAF V600E-mutant metastatic CRC patients. In this review, we elucidate the unique tumor biology of BRAF V600E-mutant CRC, with a particular focus on the immune features underlying the rationale for ICI treatments in the MSI-H and MSS subpopulations, then highlight the trends in clinical trials of the ICI therapy for BRAF V600E-mutant metastatic CRC.
Collapse
Affiliation(s)
- Mengling Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qing Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Keshu Hu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yu Dong
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xun Sun
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhiguo Zou
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Dingkun Ji
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
33
|
Underwood PW, Pawlik TM. Precision Medicine for Metastatic Colorectal Cancer: Where Do We Stand? Cancers (Basel) 2024; 16:3870. [PMID: 39594824 PMCID: PMC11593240 DOI: 10.3390/cancers16223870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Metastatic colorectal cancer is a leading cause of cancer-related death across the world. The treatment paradigm has shifted away from systemic chemotherapy alone to include targeted therapy and immunotherapy. The past two decades have been characterized by increased investigation into molecular profiling of colorectal cancer. These molecular profiles help physicians to better understand colorectal cancer biology among patients with metastatic disease. Additionally, improved data on genetic pathways allow for specific therapies to be targeted at the underlying molecular profile. Investigation of the EGFR, VEGF, HER2, and other pathways, as well as deficient mismatch repair, has led to the development of multiple targeted therapies that are now utilized in the National Comprehensive Cancer Network guidelines for colon and rectal cancer. While these new therapies have contributed to improved survival for metastatic colorectal cancer, long-term survival remains poor. Additional investigation to understand resistance to targeted therapy and development of new targeted therapy is necessary. New therapies are under development and are being tested in the preclinical and clinical settings. The aim of this review is to provide a comprehensive evaluation of molecular profiling, currently available therapies, and ongoing obstacles in the field of colorectal cancer.
Collapse
Affiliation(s)
| | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center, 395 W. 12th Ave., Suite 670, Columbus, OH 43210, USA;
| |
Collapse
|
34
|
Xie Q, Liu X, Liu R, Pan J, Liang J. Cellular mechanisms of combining innate immunity activation with PD-1/PD-L1 blockade in treatment of colorectal cancer. Mol Cancer 2024; 23:252. [PMID: 39529058 PMCID: PMC11555832 DOI: 10.1186/s12943-024-02166-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
PD-1/PD-L1 blockade therapies have displayed extraordinary clinical efficacy for melanoma, renal, bladder and lung cancer; however, only a minority of colorectal cancer (CRC) patients benefit from these treatments. The efficacy of PD-1/PD-L1 blockade in CRC is limited by the complexities of tumor microenvironment. PD-1/PD-L1 blockade immunotherapy is based on T cell-centered view of tumor immunity. However, the onset and maintenance of T cell responses and the development of long-lasting memory T cells depend on innate immune responses. Acknowledging the pivotal role of innate immunity in anti-tumor immune response, this review encapsulates the employment of combinational therapies those involve PD-1/PD-L1 blockade alongside the activation of innate immunity and explores the underlying cellular mechanisms, aiming to harnessing innate immune responses to induce long-lasting tumor control for CRC patients who received PD-1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Qi Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Xiaolin Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Rengyun Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| | - Jing Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China.
| |
Collapse
|
35
|
Zhang C, Li W, Deng M, Jiang Y, Cui X, Chen P. SIG: Graph-Based Cancer Subtype Stratification With Gene Mutation Structural Information. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:1752-1764. [PMID: 38875076 DOI: 10.1109/tcbb.2024.3414498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2024]
Abstract
Somatic tumors have a high-dimensional, sparse, and small sample size nature, making cancer subtype stratification based on somatic genomic data a challenge. Current methods for improving cancer clustering performance focus on dimension reduction, integrating multi-omics data, or generating realistic samples, yet ignore the associations between mutated genes within the patient-gene matrix. We refer to these associations as gene mutation structural information, which implicitly includes cancer subtype information and can enhance subtype clustering. We introduce a novel method for cancer subtype clustering called SIG(Structural Information within Graph). As cancer is driven by a combination of genes, we establish associations between mutated genes within the same patient sample, pair by pair, and use a graph to represent them. An association between two mutated genes corresponds to an edge in the graph. We then merge these associations among all mutated genes to obtain a structural information graph, which enriches the gene network and improves its relevance to cancer clustering. We integrate the somatic tumor genome with the enriched gene network and propagate it to cluster patients with mutations in similar network regions. Our method achieves superior clustering performance compared to SOTA methods, as demonstrated by clustering experiments on ovarian and LUAD datasets.
Collapse
|
36
|
Bollhagen A, Bodenmiller B. Highly Multiplexed Tissue Imaging in Precision Oncology and Translational Cancer Research. Cancer Discov 2024; 14:2071-2088. [PMID: 39485249 PMCID: PMC11528208 DOI: 10.1158/2159-8290.cd-23-1165] [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: 10/05/2023] [Revised: 05/24/2024] [Accepted: 08/13/2024] [Indexed: 11/03/2024]
Abstract
Precision oncology tailors treatment strategies to a patient's molecular and health data. Despite the essential clinical value of current diagnostic methods, hematoxylin and eosin morphology, immunohistochemistry, and gene panel sequencing offer an incomplete characterization. In contrast, highly multiplexed tissue imaging allows spatial analysis of dozens of markers at single-cell resolution enabling analysis of complex tumor ecosystems; thereby it has the potential to advance our understanding of cancer biology and supports drug development, biomarker discovery, and patient stratification. We describe available highly multiplexed imaging modalities, discuss their advantages and disadvantages for clinical use, and potential paths to implement these into clinical practice. Significance: This review provides guidance on how high-resolution, multiplexed tissue imaging of patient samples can be integrated into clinical workflows. It systematically compares existing and emerging technologies and outlines potential applications in the field of precision oncology, thereby bridging the ever-evolving landscape of cancer research with practical implementation possibilities of highly multiplexed tissue imaging into routine clinical practice.
Collapse
Affiliation(s)
- Alina Bollhagen
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
- Life Science Zurich Graduate School, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Bernd Bodenmiller
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
37
|
de Moraes FCA, de Oliveira Rodrigues ALS, Priantti JN, Limachi-Choque J, Burbano RMR. Efficacy and Safety of Anti-EGFR Therapy Rechallenge in Metastatic Colorectal Cancer: A Systematic Review and Meta-Analysis. J Gastrointest Cancer 2024; 56:9. [PMID: 39436445 DOI: 10.1007/s12029-024-01128-1] [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: 09/10/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) represents the second leading cause of cancer-related mortality worldwide, with a significant portion of patients presenting with metastatic disease at diagnosis. Resistance to initial anti-EGFR therapy, a key treatment for RAS wild-type metastatic CRC, remains a major challenge. This study aimed to assess the efficacy and safety of rechallenge with anti-EGFR therapy in patients with metastatic CRC who have progressed after prior treatments. METHODS A systematic search was conducted across PubMed, Web of Science, Cochrane, and Scopus. Studies were included if they were randomized controlled trials (RCTs) or observational studies involving patients with EGFR-mutated metastatic CRC who received anti-EGFR therapy as a rechallenge. Endpoints included objective response rate (ORR), disease control rate (DCR), and the incidence of adverse events. Statistical analyses were performed using the DerSimonian/Laird random effect model, with heterogeneity assessed via I2 statistics. R, version 4.2.3, was used for statistical analyses. RESULTS Fourteen studies were included with 520 patients; 50.3% were male, and the median age was 63 years old. The median progression-free survival (mPFS) ranged between 2.4 and 4.9 months, while the median overall survival (mOS) ranged from 5 to 17.8 months. Our pooled analysis demonstrated an objective response rate (ORR) of 17.70% (95% CI, 8.58-26.82%) and a disease control rate (DCR) of 61.72% (95% CI, 53.32-70.11%), both with significant heterogeneity (I2, 84% and 80%, respectively; p < 0.01). In the subgroup analysis, cetuximab showed an ORR of 18.31% (95% CI, 4.67-31.94%), and panitumumab an ORR of 10.9% (95% CI, 0.00-26.82%), while the combination of both resulted in an ORR of 29.24% (95% CI, 0.00-65.84%). For DCR, cetuximab resulted in 62.1% (95% CI, 49.32-74.87%), panitumumab in 63.05% (95% CI, 52.13-73.97%), and the combination in 60.34% (95% CI, 31.92-88.77%), all with significant heterogeneity. Adverse events included anemia (15.39%), diarrhea (4.20%), hypomagnesemia (6.40%), neutropenia (22.57%), and skin rash (13.22%). CONCLUSIONS Rechallenge with anti-EGFR therapy in metastatic CRC patients shows moderate efficacy with manageable safety profiles. These findings highlight the need for careful patient selection and monitoring to optimize outcomes. Further studies are warranted to refine strategies for maximizing the therapeutic benefits of anti-EGFR rechallenge.
Collapse
|
38
|
Li Y, Peng J, Meng X. Gut bacteria, host immunity, and colorectal cancer: From pathogenesis to therapy. Eur J Immunol 2024; 54:e2451022. [PMID: 38980275 DOI: 10.1002/eji.202451022] [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: 01/21/2024] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 07/10/2024]
Abstract
The emergence of 16S rRNA and metagenomic sequencing has gradually revealed the close relationship between dysbiosis and colorectal cancer (CRC). Recent studies have confirmed that intestinal dysbiosis plays various roles in the occurrence, development, and therapeutic response of CRC. Perturbation of host immunity is one of the key mechanisms involved. The intestinal microbiota, or specific bacteria and their metabolites, can modulate the progression of CRC through pathogen recognition receptor signaling or via the recruitment, polarization, and activation of both innate and adaptive immune cells to reshape the protumor/antitumor microenvironment. Therefore, the administration of gut bacteria to enhance immune homeostasis represents a new strategy for the treatment of CRC. In this review, we cover recent studies that illuminate the role of gut bacteria in the progression and treatment of CRC through orchestrating the immune response, which potentially offers insights for subsequent transformative research.
Collapse
Affiliation(s)
- Yuyi Li
- Department of Gastroenterology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Gut Microecology and Associated Major Diseases Research, Shanghai, China
- Digestive Disease Research and Clinical Translation Center, Shanghai Jiao Tong University, Shanghai, China
| | - Jinjin Peng
- Department of Gastroenterology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Gut Microecology and Associated Major Diseases Research, Shanghai, China
- Digestive Disease Research and Clinical Translation Center, Shanghai Jiao Tong University, Shanghai, China
| | - Xiangjun Meng
- Department of Gastroenterology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Gut Microecology and Associated Major Diseases Research, Shanghai, China
- Digestive Disease Research and Clinical Translation Center, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
39
|
Maier JA, Castiglioni S, Petrelli A, Cannatelli R, Ferretti F, Pellegrino G, Sarzi Puttini P, Fiorina P, Ardizzone S. Immune-Mediated Inflammatory Diseases and Cancer - a dangerous liaison. Front Immunol 2024; 15:1436581. [PMID: 39359726 PMCID: PMC11445042 DOI: 10.3389/fimmu.2024.1436581] [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: 05/22/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
Patients with Immune-Mediated Inflammatory Diseases (IMIDs) are known to have an elevated risk of developing cancer, but the exact causative factors remain subject to ongoing debate. This narrative review aims to present the available evidence concerning the intricate relationship between these two conditions. Environmental influences and genetic predisposition lead to a dysregulated immune response resulting in chronic inflammation, which is crucial in the pathogenesis of IMIDs and oncogenic processes. Mechanisms such as the inflammatory microenvironment, aberrant intercellular communication due to abnormal cytokine levels, excessive reparative responses, and pathological angiogenesis are involved. The chronic immunosuppression resulting from IMIDs treatments further adds to the complexity of the pathogenic scenario. In conclusion, this review highlights critical gaps in the current literature, suggesting potential avenues for future research. The intricate interplay between IMIDs and cancer necessitates more investigation to deepen our understanding and improve patient management.
Collapse
Affiliation(s)
- Jeanette A Maier
- Department of Biomedical and Clinical Sciences, Università di Milano, Milano, Italy
| | - Sara Castiglioni
- Department of Biomedical and Clinical Sciences, Università di Milano, Milano, Italy
| | - Alessandra Petrelli
- Department of Clinical Sciences and Community Health, University of Milan, Milano, Italy
| | | | | | | | - Piercarlo Sarzi Puttini
- Department of Biomedical and Clinical Sciences, Università di Milano, Milano, Italy
- IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milano, Italy
| | - Paolo Fiorina
- Department of Biomedical and Clinical Sciences, Università di Milano, Milano, Italy
| | - Sandro Ardizzone
- Gastroenterology Unit, ASST Fatebenefratelli-Sacco, Milano, Italy
| |
Collapse
|
40
|
Benboubker V, Ramzy GM, Jacobs S, Nowak-Sliwinska P. Challenges in validation of combination treatment strategies for CRC using patient-derived organoids. J Exp Clin Cancer Res 2024; 43:259. [PMID: 39261955 PMCID: PMC11389238 DOI: 10.1186/s13046-024-03173-x] [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: 07/17/2024] [Accepted: 08/23/2024] [Indexed: 09/13/2024] Open
Abstract
Patient-derived organoids (PDOs) established from tissues from various tumor types gave the foundation of ex vivo models to screen and/or validate the activity of many cancer drug candidates. Due to their phenotypic and genotypic similarity to the tumor of which they were derived, PDOs offer results that effectively complement those obtained from more complex models. Yet, their potential for predicting sensitivity to combination therapy remains underexplored. In this review, we discuss the use of PDOs in both validation and optimization of multi-drug combinations for personalized treatment strategies in CRC. Moreover, we present recent advancements in enriching PDOs with diverse cell types, enhancing their ability to mimic the complexity of in vivo environments. Finally, we debate how such sophisticated models are narrowing the gap in personalized medicine, particularly through immunotherapy strategies and discuss the challenges and future direction in this promising field.
Collapse
Affiliation(s)
- Valentin Benboubker
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel-Servet, Geneva, 4 1211, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, 1211, Switzerland
- Translational Research Center in Oncohaematology, Geneva, 1211, Switzerland
| | - George M Ramzy
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel-Servet, Geneva, 4 1211, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, 1211, Switzerland
- Translational Research Center in Oncohaematology, Geneva, 1211, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, 1211, Switzerland
| | - Sacha Jacobs
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel-Servet, Geneva, 4 1211, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, 1211, Switzerland
- Translational Research Center in Oncohaematology, Geneva, 1211, Switzerland
| | - Patrycja Nowak-Sliwinska
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel-Servet, Geneva, 4 1211, Switzerland.
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, 1211, Switzerland.
- Translational Research Center in Oncohaematology, Geneva, 1211, Switzerland.
| |
Collapse
|
41
|
Yu Y, Wu H, Hong L, Qiu J, Wu S, Shao L, Lin C, Wang Z, Wu J. A large population-based and validated study on the follow-up management and supportive strategy of locally advanced rectal cancer patients. Support Care Cancer 2024; 32:652. [PMID: 39256234 DOI: 10.1007/s00520-024-08860-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/05/2024] [Indexed: 09/12/2024]
Abstract
OBJECTIVE Our objective was to evaluate the predictive factors and metastatic time for liver and lung metastasis in locally advanced rectal cancer (RC) patients. METHODS Univariate and multivariate analysis were performed to identify risk factors and prognostic factors for liver metastasis and lung metastasis in RC. Survival probabilities were calculated using the Kaplan-Meier model and compared using the log-rank test between groups. The probability of time-to-event occurrence was calculated using the random survival forest model. Finally, the SEER database was used to verify our findings. RESULTS Our results indicated that pathological T stage and pathological N stage were independent predictive factors for liver metastasis. Furthermore, CEA level, pathological T stage, and tumor deposit were independent predictive factors for lung metastasis. Based on the results of a multivariate Cox analysis, we categorized patients with liver and lung metastasis into three groups based on their scores. The results revealed that patients with higher scores had a higher probability of experiencing metastasis. For liver metastasis, Groups 1, 2, and 3 all exhibited higher occurrence rates within the first 24 months. However, for lung metastasis, Group 4 showed the highest occurrence rate at the 12th month, while Groups 5 and 6 exhibited the highest occurrence rates at the 15th month. CONCLUSIONS In summary, we developed predictive models to determine the likelihood of liver and lung metastasis in RC patients. It is crucial to implement a more intensive surveillance program for patients with unfavorable risk profiles in order to facilitate early detection of metastasis.
Collapse
Affiliation(s)
- Yilin Yu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Haixia Wu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Liang Hong
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Jianjian Qiu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Shiji Wu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Lingdong Shao
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Cheng Lin
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China.
| | - Zhiping Wang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China.
| | - Junxin Wu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China.
| |
Collapse
|
42
|
Chu X, Li X, Zhang Y, Dang G, Miao Y, Xu W, Wang J, Zhang Z, Cheng S. Integrative single-cell analysis of human colorectal cancer reveals patient stratification with distinct immune evasion mechanisms. NATURE CANCER 2024; 5:1409-1426. [PMID: 39147986 DOI: 10.1038/s43018-024-00807-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 07/16/2024] [Indexed: 08/17/2024]
Abstract
The tumor microenvironment (TME) considerably influences colorectal cancer (CRC) progression, therapeutic response and clinical outcome, but studies of interindividual heterogeneities of the TME in CRC are lacking. Here, by integrating human colorectal single-cell transcriptomic data from approximately 200 donors, we comprehensively characterized transcriptional remodeling in the TME compared to noncancer tissues and identified a rare tumor-specific subset of endothelial cells with T cell recruitment potential. The large sample size enabled us to stratify patients based on their TME heterogeneity, revealing divergent TME subtypes in which cancer cells exploit different immune evasion mechanisms. Additionally, by associating single-cell transcriptional profiling with risk genes identified by genome-wide association studies, we determined that stromal cells are major effector cell types in CRC genetic susceptibility. In summary, our results provide valuable insights into CRC pathogenesis and might help with the development of personalized immune therapies.
Collapse
Affiliation(s)
| | | | - Yu Zhang
- Changping Laboratory, Beijing, China
| | - Guohui Dang
- Changping Laboratory, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | | | - Wenbin Xu
- Changping Laboratory, Beijing, China
| | | | - Zemin Zhang
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| | | |
Collapse
|
43
|
Tian S, Chen M. The mechanisms and drug therapies of colorectal cancer and epigenetics: bibliometrics and visualized analysis. Front Pharmacol 2024; 15:1466156. [PMID: 39268463 PMCID: PMC11391208 DOI: 10.3389/fphar.2024.1466156] [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: 07/17/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Numerous studies have demonstrated a link between epigenetics and CRC. However, there has been no systematic analysis or visualization of relevant publications using bibliometrics. METHODS 839 publications obtained from the Web of Science Core (WoSCC) were systematically analyzed using CiteSpace and VOSviewer software. RESULTS The results show that the countries, institutions, and authors with the most published articles are the United States, Harvard University, and Ogino and Shuji, respectively. SEPT9 is a blood test for the early detection of colorectal cancer. Vitamin D and gut microbiota mediate colorectal cancer and epigenetics, and probiotics may reduce colorectal cancer-related symptoms. We summarize the specific epigenetic mechanisms of CRC and the current existence and potential epigenetic drugs associated with these mechanisms. It is closely integrated with clinical practice, and the possible research directions and challenges in the future are proposed. CONCLUSION This study reviews the current research trends and hotspots in CRC and epigenetics, which can promote the development of this field and provide references for researchers in this field.
Collapse
Affiliation(s)
- Siyu Tian
- School of Clinical Medicine, Chengdu University of TCM, Chengdu, China
| | - Min Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
44
|
Reitsam NG, Grosser B, Steiner DF, Grozdanov V, Wulczyn E, L'Imperio V, Plass M, Müller H, Zatloukal K, Muti HS, Kather JN, Märkl B. Converging deep learning and human-observed tumor-adipocyte interaction as a biomarker in colorectal cancer. COMMUNICATIONS MEDICINE 2024; 4:163. [PMID: 39147895 PMCID: PMC11327259 DOI: 10.1038/s43856-024-00589-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 08/05/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Tumor-Adipose-Feature (TAF) as well as SARIFA (Stroma AReactive Invasion Front Areas) are two histologic features/biomarkers linking tumor-associated adipocytes to poor outcomes in colorectal cancer (CRC) patients. Whereas TAF was identified by deep learning (DL) algorithms, SARIFA was established as a human-observed histopathologic biomarker. METHODS To study the overlap between TAF and SARIFA, we performed a systematic pathological review of TAF based on all published image tiles. Additionally, we analyzed the presence/absence of TAF in SARIFA-negative CRC cases to elucidate the biologic and prognostic role of a direct tumor-adipocyte contact. TCGA-CRC gene expression data is investigated to assess the association of FABP4 (fatty-acid binding protein 4) and CD36 (fatty-acid translocase) with both TAF and CRC prognosis. RESULTS By investigating the TAF/SARIFA overlap, we show that many TAF patches correspond to the recently described SARIFA-phenomenon. Even though there is a pronounced morphological and biological overlap, there are differences in the concepts. The presence of TAF in SARIFA-negative CRCs is not associated with poor outcomes in this cohort, potentially highlighting the importance of a direct tumor-adipocyte interaction. Upregulation of FABP4 and CD36 gene expression seem both linked to a poor prognosis in CRC. CONCLUSIONS By proving the substantial overlap between human-observed SARIFA and DL-based TAF as morphologic biomarkers, we demonstrate that linking DL-based image features to independently developed histopathologic biomarkers is a promising tool in the identification of clinically and biologically meaningful biomarkers. Adipocyte-tumor-cell interactions seem to be crucial in CRC, which should be considered as biomarkers for further investigations.
Collapse
Affiliation(s)
- Nic G Reitsam
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany.
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany.
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany.
| | - Bianca Grosser
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | | | | | - Ellery Wulczyn
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Vincenzo L'Imperio
- Department of Medicine and Surgery, Pathology, University of Milano-Bicocca, IRCCS (Scientific Institute for Research, Hospitalization and Healthcare) Fondazione San Gerardo dei Tintori, Monza, Italy
| | - Markus Plass
- Medical University of Graz, Diagnostic and Research Institute of Pathology, Graz, Austria
| | - Heimo Müller
- Medical University of Graz, Diagnostic and Research Institute of Pathology, Graz, Austria
| | - Kurt Zatloukal
- Medical University of Graz, Diagnostic and Research Institute of Pathology, Graz, Austria
| | - Hannah S Muti
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Jakob N Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
- Department of Medicine I, University Hospital Dresden, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Bruno Märkl
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| |
Collapse
|
45
|
Lee S, Kim S, Koh G, Ahn H. Identification of Time-Series Pattern Marker in Its Application to Mortality Analysis of Pneumonia Patients in Intensive Care Unit. J Pers Med 2024; 14:812. [PMID: 39202004 PMCID: PMC11355743 DOI: 10.3390/jpm14080812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Electronic Health Records (EHRs) are a significant source of big data used to track health variables over time. The analysis of EHR data can uncover medical markers or risk factors, aiding in the diagnosis and monitoring of diseases. We introduce a novel method for identifying markers with various temporal trend patterns, including monotonic and fluctuating trends, using machine learning models such as Long Short-Term Memory (LSTM). By applying our method to pneumonia patients in the intensive care unit using the MIMIC-III dataset, we identified markers exhibiting both monotonic and fluctuating trends. Specifically, monotonic markers such as red cell distribution width, urea nitrogen, creatinine, calcium, morphine sulfate, bicarbonate, sodium, troponin T, albumin, and prothrombin time were more frequently observed in the mortality group compared to the recovery group throughout the 10-day period before discharge. Conversely, fluctuating trend markers such as dextrose in sterile water, polystyrene sulfonate, free calcium, and glucose were more frequently observed in the mortality group as the discharge date approached. Our study presents a method for detecting time-series pattern markers in EHR data that respond differently according to disease progression. These markers can contribute to monitoring disease progression and enable stage-specific treatment, thereby advancing precision medicine.
Collapse
Affiliation(s)
- Suhyeon Lee
- Division of Data Science, The University of Suwon, Hwaseong-si 16419, Republic of Korea; (S.L.); (S.K.); (G.K.)
- DS&ML Center, The University of Suwon, Hwaseong-si 16419, Republic of Korea
| | - Suhyun Kim
- Division of Data Science, The University of Suwon, Hwaseong-si 16419, Republic of Korea; (S.L.); (S.K.); (G.K.)
- DS&ML Center, The University of Suwon, Hwaseong-si 16419, Republic of Korea
| | - Gayoun Koh
- Division of Data Science, The University of Suwon, Hwaseong-si 16419, Republic of Korea; (S.L.); (S.K.); (G.K.)
- DS&ML Center, The University of Suwon, Hwaseong-si 16419, Republic of Korea
| | - Hongryul Ahn
- Division of Data Science, The University of Suwon, Hwaseong-si 16419, Republic of Korea; (S.L.); (S.K.); (G.K.)
- DS&ML Center, The University of Suwon, Hwaseong-si 16419, Republic of Korea
| |
Collapse
|
46
|
van der Graaff D, Seghers S, Vanclooster P, Deben C, Vandamme T, Prenen H. Advancements in Research and Treatment Applications of Patient-Derived Tumor Organoids in Colorectal Cancer. Cancers (Basel) 2024; 16:2671. [PMID: 39123399 PMCID: PMC11311786 DOI: 10.3390/cancers16152671] [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: 07/05/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Colorectal cancer (CRC) remains a significant health burden globally, being the second leading cause of cancer-related mortality. Despite significant therapeutic advancements, resistance to systemic antineoplastic agents remains an important obstacle, highlighting the need for innovative screening tools to tailor patient-specific treatment. This review explores the application of patient-derived tumor organoids (PDTOs), three-dimensional, self-organizing models derived from patient tumor samples, as screening tools for drug resistance in CRC. PDTOs offer unique advantages over traditional models by recapitulating the tumor architecture, cellular heterogeneity, and genomic landscape and are a valuable ex vivo predictive drug screening tool. This review provides an overview of the current literature surrounding the use of PDTOs as an instrument for predicting therapy responses in CRC. We also explore more complex models, such as co-cultures with important stromal cells, such as cancer-associated fibroblasts, and organ-on-a-chip models. Furthermore, we discuss the use of PDTOs for drug repurposing, offering a new approach to identify the existing drugs effective against drug-resistant CRC. Additionally, we explore how PDTOs serve as models to gain insights into drug resistance mechanisms, using newer techniques, such as single-cell RNA sequencing and CRISPR-Cas9 genome editing. Through this review, we aim to highlight the potential of PDTOs in advancing our understanding of predicting therapy responses, drug resistance, and biomarker identification in CRC management.
Collapse
Affiliation(s)
| | - Sofie Seghers
- Department of Medical Oncology, University Hospital Antwerp, 2650 Edegem, Belgium
- Center for Oncological Research (CORE), University of Antwerp, 2610 Wilrijk, Belgium
| | | | - Christophe Deben
- Center for Oncological Research (CORE), University of Antwerp, 2610 Wilrijk, Belgium
| | - Timon Vandamme
- Department of Medical Oncology, University Hospital Antwerp, 2650 Edegem, Belgium
- Center for Oncological Research (CORE), University of Antwerp, 2610 Wilrijk, Belgium
| | - Hans Prenen
- Department of Medical Oncology, University Hospital Antwerp, 2650 Edegem, Belgium
- Center for Oncological Research (CORE), University of Antwerp, 2610 Wilrijk, Belgium
| |
Collapse
|
47
|
Gong Z, Huang X, Cao Q, Wu Y, Zhang Q. A CLRN3-Based CD8 + T-Related Gene Signature Predicts Prognosis and Immunotherapy Response in Colorectal Cancer. Biomolecules 2024; 14:891. [PMID: 39199281 PMCID: PMC11352867 DOI: 10.3390/biom14080891] [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: 04/24/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) ranks among the most prevalent malignancies affecting the gastrointestinal tract. The infiltration of CD8+ T cells significantly influences the prognosis and progression of tumor patients. METHODS This study establishes a CRC immune risk model based on CD8+ T cell-related genes. CD8+ T cell-related genes were identified through Weighted Gene Co-expression Network Analysis (WGCNA), and the enriched gene sets were annotated via Gene Ontology (GO) and Reactome pathway analysis. Employing machine learning methods, including the Least Absolute Shrinkage and Selection Operator (LASSO) algorithm and Random Forest (RF), we identified nine genes associated with CD8+ T-cell infiltration. The infiltration levels of immune cells in CRC tissues were assessed using the ssGSEA algorithm. RESULTS These genes provide a foundation for constructing a prognostic model. The TCGA-CRC sample model's prediction scores were categorized, and the prediction models were validated through Cox regression analysis and Kaplan-Meier curve analysis. Notably, although CRC tissues with higher risk scores exhibited elevated levels of CD8+ T-cell infiltration, they also demonstrated heightened expression of immune checkpoint genes. Furthermore, comparison of microsatellite instability (MSI) and gene mutations across the immune subgroups revealed notable gene variations, particularly with APC, TP53, and TNNT1 showing higher mutation frequencies. Finally, the predictive model's efficacy was corroborated through the use of Tumor Immune Dysfunction and Exclusion (TIDE), Immune Profiling Score (IPS), and immune escape-related molecular markers. The predictive model was validated through an external cohort of CRC and the Bladder Cancer Immunotherapy Cohort. CLRN3 expression levels in tumor and adjacent normal tissues were assessed using quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. Subsequent in vitro and in vivo experiments demonstrated that CLRN3 knockdown significantly attenuated the malignant biological behavior of CRC cells, while overexpression had the opposite effect. CONCLUSIONS This study presents a novel prognostic model for CRC, providing a framework for enhancing the survival rates of CRC patients by targeting CD8+ T-cell infiltration.
Collapse
Affiliation(s)
- Zhiwen Gong
- Department of Thoracic Surgery, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China; (Z.G.); (Q.C.)
| | - Xiuting Huang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China;
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China
| | - Qingdong Cao
- Department of Thoracic Surgery, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China; (Z.G.); (Q.C.)
| | - Yuanquan Wu
- Department of Gastrointestinal Surgery, The Affiliated Kashi Hospital, Sun Yat-Sen University, Kashi 844000, China
| | - Qunying Zhang
- Department of Geriatrics, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, China
| |
Collapse
|
48
|
Napolitano S, Martini G, Ciardiello D, Del Tufo S, Martinelli E, Troiani T, Ciardiello F. Targeting the EGFR signalling pathway in metastatic colorectal cancer. Lancet Gastroenterol Hepatol 2024; 9:664-676. [PMID: 38697174 DOI: 10.1016/s2468-1253(23)00479-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/21/2023] [Accepted: 12/28/2023] [Indexed: 05/04/2024]
Abstract
Epidermal growth factor receptor (EGFR) and its activated downstream signalling pathways play a crucial role in colorectal cancer development and progression. After four decades of preclinical, translational, and clinical research, it has been shown that blocking the EGFR signalling pathway at different molecular levels represents a fundamental therapeutic strategy for patients with metastatic colorectal cancer. Nevertheless, the efficacy of molecularly targeted therapies is inescapably limited by the insurgence of mechanisms of acquired cancer cell resistance. Thus, in the era of precision medicine, a deeper understanding of the complex molecular landscape of metastatic colorectal cancer is required to deliver the best treatment choices to all patients. Major efforts are currently ongoing to improve patient selection, improve the efficacy of available treatments targeting the EGFR pathway, and develop novel combination strategies to overcome therapy resistance within the continuum of care of metastatic colorectal cancer.
Collapse
Affiliation(s)
- Stefania Napolitano
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Giulia Martini
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Davide Ciardiello
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy; Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | - Sara Del Tufo
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Erika Martinelli
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Teresa Troiani
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università degli studi della Campania Luigi Vanvitelli, Napoli, Italy.
| |
Collapse
|
49
|
Dang Q, Zuo L, Hu X, Zhou Z, Chen S, Liu S, Ba Y, Zuo A, Xu H, Weng S, Zhang Y, Luo P, Cheng Q, Liu Z, Han X. Molecular subtypes of colorectal cancer in the era of precision oncotherapy: Current inspirations and future challenges. Cancer Med 2024; 13:e70041. [PMID: 39054866 PMCID: PMC11272957 DOI: 10.1002/cam4.70041] [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: 03/29/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is among the most hackneyed malignancies. Even patients with identical clinical symptoms and the same TNM stage still exhibit radically different clinical outcomes after receiving equivalent treatment regimens, indicating extensive heterogeneity of CRC. Myriad molecular subtypes of CRC have been exploited for decades, including the most compelling consensus molecular subtype (CMS) classification that has been broadly applied for patient stratification and biomarker-drug combination formulation. Encountering barriers to clinical translation, however, CMS classification fails to fully reflect inter- or intra-tumor heterogeneity of CRC. As a consequence, addressing heterogeneity and precisely managing CRC patients with unique characteristics remain arduous tasks for clinicians. REVIEW In this review, we systematically summarize molecular subtypes of CRC and further elaborate on their clinical applications, limitations, and future orientations. CONCLUSION In recent years, exploration of subtypes through cell lines, animal models, patient-derived xenografts (PDXs), organoids, and clinical trials contributes to refining biological insights and unraveling subtype-specific therapies in CRC. Therapeutic interventions including nanotechnology, clustered regulatory interspaced short palindromic repeat/CRISPR-associated nuclease 9 (CRISPR/Cas9), gut microbiome, and liquid biopsy are powerful tools with the possibility to shift the immunologic landscape and outlook for CRC precise medicine.
Collapse
Affiliation(s)
- Qin Dang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Lulu Zuo
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xinru Hu
- Department of Cardiology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Zhaokai Zhou
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shuang Chen
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shutong Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuhao Ba
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Anning Zuo
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hui Xu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Siyuan Weng
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuyuan Zhang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Peng Luo
- Department of Oncology, Zhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Quan Cheng
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Zaoqu Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
- Institute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xinwei Han
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
50
|
Li Y, Li H, Sun G, Xu S, Tang X, Zhang L, Wan L, Zhang L, Tang M. Integrative analyses of multi-omics data constructing tumor microenvironment and immune-related molecular prognosis model in human colorectal cancer. Heliyon 2024; 10:e32744. [PMID: 38975206 PMCID: PMC11226854 DOI: 10.1016/j.heliyon.2024.e32744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 07/09/2024] Open
Abstract
The increasing prevalence and incidence of colorectal cancer (CRC), particularly in young adults, underscore the imperative to comprehend its fundamental mechanisms, discover novel diagnostic and prognostic markers, and enhance therapeutic strategies. Here, we integrated multi-omics data, including gene expression, somatic mutation data and DNA methylation data, to unravel the intricacies of tumor microenvironment (TME) in CRC and search for novel prognostic markers. By calculating the immune score for each patient from the expression profile, we delineated the differential immune cell fraction, constructed an immune-related multi-omics atlas, and identified molecular characteristics. The entire colorectal dataset (n = 343) was randomly divided into training (n = 249) and testing datasets (n = 94). We screened 144 immune-related genes, 6 mutant genes, and 38 methylation probes associated with overall survival (OS). These makers were then incorporated into a 10-gene prognostic model using Lasso and Cox regression in the training dataset, and the model's performance was evaluated in an independent validation dataset. The model exhibited satisfactory results (average concordance index [C-index] = 0.77), with the average 1-year, 3-year, and 5-year AUCs being 0.79, 0.76, and 0.76 in the training dataset and 0.74, 0.80, and 0.90 in the testing dataset. Furthermore, the prognostic model demonstrated applicability in guiding chemotherapy for CRC patients and exhibited a degree of pan-cancer utility in risk stratification. In conclusion, our integrated analysis of multi-omics data revealed immune-related genetic and epigenetic characteristics of the TME. We propose an integrative prognostic model that can stratify risk and guide chemotherapy for CRC patients. The generalizability of the model in risk stratification across different cancer types was validated in Pan-Cancer cohort.
Collapse
Affiliation(s)
- Yifei Li
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Hexin Li
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Gaoyuan Sun
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Siyuan Xu
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiaokun Tang
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Lanxin Zhang
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Li Wan
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Lili Zhang
- Clinical Biobank, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Min Tang
- Department of Medical Oncology, Institute of Geriatric Medicine, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|