1
|
Yang J, Li X, Li T, Mei J, Chen Y. Recent advances in biomimetic nanodelivery systems for cancer Immunotherapy. Mater Today Bio 2025; 32:101726. [PMID: 40270890 PMCID: PMC12017925 DOI: 10.1016/j.mtbio.2025.101726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/26/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
Tumor immunotherapy is a developing and promising therapeutic method. However, the mechanism of tumor immune microenvironment and individual differences of patients make the clinical application of immunotherapy still very limited. The resulting targeting of the tumor environment and immune system is a suitable strategy for tumor therapy. Biomimetic nanodelivery systems (BNDS) coated with nanoparticles has brought new hope for tumor immunotherapy. Due to its high targeting, maximum drug delivery efficiency and immune escape, BNDS has become one of the options for tumor immunotherapy in the future. BNDS combines the advantages of natural cell membranes and nanoparticles and has good targeting properties. This review summarizes the relationship between tumor and immune microenvironment, classification of immunotherapy, engineering modification of cell membrane, and a comprehensive overview of different types of membrane BNDS in immunotherapy. Furthermore, the prospects and challenges of biomimetic nanoparticles coated with membranes in tumor immunotherapy are further discussed.
Collapse
Affiliation(s)
- Jiawei Yang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China, No. 508 North Second Ring East Road, Ningbo, 315302, Zhejiang, China
| | - Xueqi Li
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China, No. 508 North Second Ring East Road, Ningbo, 315302, Zhejiang, China
| | - Tongyu Li
- Department of Hematology, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, 315010, China
| | - Jin Mei
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China, No. 508 North Second Ring East Road, Ningbo, 315302, Zhejiang, China
- Institute of Engineering Medicine, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, 315010, China
| | - Ying Chen
- Institute of Engineering Medicine, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, 315010, China
| |
Collapse
|
2
|
Jain M, Jadhav IM, Dangat SV, Singuru SR, Sethi G, Yuba E, Gupta RK. Overcoming the novel glycan-lectin checkpoints in tumor microenvironments for the success of the cross-presentation-based immunotherapy. Biomater Sci 2025. [PMID: 40421610 DOI: 10.1039/d4bm01732c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
In pursuit of meeting the ever-rising demand for cancer therapies, cross-presentation-based glyconanovaccines (GNVs) targeting C-type lectin receptors (CLRs) on DCs have shown significant potential as cutting-edge cancer immunotherapy. GNVs are an attractive approach to induce anti-cancer cytotoxic T lymphocyte responses. Despite immune checkpoints (ICs) being well established and an obstacle to the success of GNVs, glycan-lectin circuits are emerging as unique checkpoints due to their immunomodulatory functions. Given the role of aberrant tumor glycosylation in promoting immune evasion, mitigating these effects is crucial for the efficacy of GNVs. Lectins, such as siglecs and galectins, are detrimental to the tumor immune landscape as they promote an immunosuppressive TME. From this perspective, this review aims to explore glycan-lectin ICs and their influence on the efficacy of GNVs. We aim to discuss various ICs in the TME followed by drawbacks of immune checkpoint inhibitors (ICIs). We will also emphasize the altered glycosylation profile of tumors, addressing their immunosuppressive nature along with ways in which CLRs, siglecs, and galectins contribute to immune evasion and cancer progression. Considering the resistance towards ICIs, current and prospective approaches for targeting glycan-lectin circuits and future prospects of these endeavors in harnessing the full potential of GNVs will also be highlighted.
Collapse
Affiliation(s)
- Mannat Jain
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Isha M Jadhav
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Suyash Vinayak Dangat
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Srinivasa Rao Singuru
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore.
| | - Eiji Yuba
- Department of Chemistry & Bioengineering, Graduate School of Engineering, Osaka Metropolitan University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka-city, Osaka 558-8585, Japan.
| | - Rajesh Kumar Gupta
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| |
Collapse
|
3
|
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
|
4
|
Tian T, Han H, Huang J, Ma J, Ran R. DBI as a Novel Immunotherapeutic Candidate in Colorectal Cancer: Dissecting Genetic Risk and the Immune Landscape via GWAS, eQTL, and pQTL. Biomedicines 2025; 13:1115. [PMID: 40426943 PMCID: PMC12109284 DOI: 10.3390/biomedicines13051115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/24/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide. Identifying drug targets associated with CRC is crucial for developing targeted therapies. Methods: MR (IVW, Wald ratio, weighted median, and MR-Egger) and SMR analyses were used to screen candidate genes associated with CRC risk. Further validation was performed using The Cancer Genome Atlas (TCGA) to assess gene expression patterns and prognostic significance. Additionally, immune infiltration analysis was conducted to characterize the tumor immune microenvironment. Drug prediction was performed to explore potential therapeutic interventions. Results: Eight genes were identified associated with CRC. IGFBP3, CD72, SERPINH1, CHRDL2, LRP11, and SPARCL1 were linked to an increased risk of CRC, whereas DBI and HYAL1 were associated with a decreased risk of CRC. Notably, DBI exhibited a potentially favorable immune profile, negatively correlated with Tregs and MDSCs while positively associated with activated CD8+ and CD4+ T cells. Conclusions: Eight genes were identified as associated with CRC, among which DBI exhibited a potential protective role, correlating with improved patient survival, enhanced immune activation, and increased responsiveness to immunotherapy. The remaining proteins demonstrated diverse and complex functions within the tumor immune microenvironment, providing novel insights for the development of precision diagnostics and immunotherapeutic strategies.
Collapse
Affiliation(s)
- Ting Tian
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China; (T.T.); (J.M.)
| | - Huan Han
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; (H.H.); (J.H.)
| | - Jingtao Huang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; (H.H.); (J.H.)
| | - Jun’e Ma
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China; (T.T.); (J.M.)
| | - Ruoxi Ran
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China; (T.T.); (J.M.)
| |
Collapse
|
5
|
Zhang Y, Xie Y, Xia S, Ge X, Li J, Liu F, Jia F, Wang S, Zhou Q, Gao M, Fang W, Zheng C. The Novel Dual GIP and GLP-1 Receptor Agonist Tirzepatide Attenuates Colon Cancer Development by Regulating Glucose Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411980. [PMID: 40125821 PMCID: PMC12097124 DOI: 10.1002/advs.202411980] [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] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 03/10/2025] [Indexed: 03/25/2025]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer mortality while diabetes is a recognized risk factor for CRC. Here we report that tirzepatide (TZP), a novel polypeptide/glucagon-like peptide 1 receptor (GIPR/GLP-1R) agonist for the treatment of diabetes, has a role in attenuating CRC growth. TZP significantly inhibited colon cancer cell proliferation promoted apoptosis in vitro and induced durable tumor regression in vivo under hyperglycemic and nonhyperglycemic conditions across multiple murine cancer models. As glucose metabolism is known to critically regulate colon cancer progression, spatial metabolomics results revealed that glucose metabolites are robustly reduced in the colon cancer regions of the TZP-treated mice. TZP inhibited glucose uptake and destabilized hypoxia-inducible factor-1 alpha (HIF-1α) with reduced expression and activity of the rate-limiting enzymes 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) and phosphofructokinase 1 (PFK-1). These effects contributed to the downregulation of glycolysis and the tricarboxylic acid (TCA) cycle. TZP also delayed tumor development in a patient-derived xenograft (PDX) mouse model accompanied by HIF-1α mediated PFKFB3-PFK-1 inhibition. Therefore, the study provides strong evidence that glycolysis-blocking TZP, besides its application in treating type 2 diabetes, has the potential for preclinical studies as a therapy for colorectal cancer used either as monotherapy or in combination with other anticancer therapies.
Collapse
Affiliation(s)
- Yikai Zhang
- Department of EndocrinologyThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Yi Xie
- Department of EndocrinologyThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Shenglong Xia
- Department of GastroenterologyThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Xinnuo Ge
- Department of EndocrinologyThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Jiaying Li
- Center for Basic and Translational ResearchThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Fang Liu
- Department of EndocrinologyThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Fan Jia
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of EducationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhou310009P. R. China
| | - Shengyao Wang
- Department of EndocrinologyThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Qiao Zhou
- Department of EndocrinologyThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Menghan Gao
- Department of EndocrinologyThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Weihuan Fang
- Department of Veterinary MedicineZhejiang UniversityHangzhou310009P. R. China
| | - Chao Zheng
- Department of EndocrinologyThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhou310009P. R. China
| |
Collapse
|
6
|
Wang C, Chen M, Chen P, Han J, Hu H, Chen J, Wu Q, Zhao D, Wang T, Zhou J, Li Q, Zhou R, Wen Y, Yang J, Shi M, Wang Y. RBM15-mediated metabolic reprogramming boosts immune response in colorectal cancer. Front Immunol 2025; 16:1515568. [PMID: 40370450 PMCID: PMC12075365 DOI: 10.3389/fimmu.2025.1515568] [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: 10/23/2024] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Introduction Immune checkpoint blockade (ICB) therapy has shown promise in treating advanced colorectal cancer, particularly in patients with microsatellite instability-high (MSI-H) tumors. However, only a subset of these patients responds favorably, highlighting the need for strategies to improve immunotherapy efficacy. Methods To identify potential regulators of immunotherapy response, we conducted a comprehensive analysis of colorectal cancer datasets from The Cancer Genome Atlas (TCGA). We performed multi-omics analyses and functional assays in both human and murine colorectal cancer cell lines. Additionally, we evaluated tumor growth and immune cell infiltration using syngeneic mouse models. Results Our analysis revealed that RNA binding motif protein 15 (RBM15) is highly expressed in colorectal cancer and correlates with poor patient prognosis. Functional studies demonstrated that RBM15 loss led to increased expression of fumarate hydratase (FH). This led to decreased levels of fumarate, a metabolite known to suppress anti-tumor immune responses. In vivo, RBM15 depletion significantly delayed tumor progression and enhanced CD8⁺ T cell infiltration and activation in the tumor microenvironment. Discussion These findings identify RBM15 as a negative regulator of anti-tumor immunity in colorectal cancer. Targeting RBM15 may represent a novel strategy to boost immune responsiveness and improve outcomes for patients undergoing immunotherapy.
Collapse
Affiliation(s)
- Chen Wang
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengyan Chen
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Panyu Chen
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jinlu Han
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Hu
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiong Chen
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiong Wu
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - De Zhao
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tongshuai Wang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyi Zhou
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Li
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runkai Zhou
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yugang Wen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Yang
- Department of Pathology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Shi
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yugang Wang
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Allam AA, Rudayni HA, Ahmed NA, Aba Alkhayl FF, Lamsabhi AM, Kamel EM. Comprehensive insights into carbonic anhydrase inhibition: A triad of In vitro, In silico, and In vivo perspectives. Enzyme Microb Technol 2025; 189:110657. [PMID: 40252302 DOI: 10.1016/j.enzmictec.2025.110657] [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: 03/13/2025] [Revised: 04/08/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
Carbonic anhydrases (CAs) are zinc-dependent metalloenzymes essential for sustaining physiological balance by facilitating the reversible conversion of carbon dioxide to its hydrated form. Their biological significance, coupled with their involvement in a wide array of pathological conditions, makes them attractive targets for therapeutic intervention. This review presents a comprehensive analysis of carbonic anhydrase inhibition through an integrated triad of in vitro, In silico, and In vivo perspectives. In vitro studies provide critical insights into the mechanisms of enzyme inhibition, enabling the identification and optimization of potent inhibitors while elucidating their structure-activity relationships. In silico methodologies, including docking, molecular dynamics (MD) simulation, virtual screening, ADMET, and QSAR analyses, have emerged as invaluable tools in rational drug design, streamlining the discovery and development of isoform-specific inhibitors. Complementing these efforts, In vivo investigations validate the pharmacokinetics, pharmacodynamics, and therapeutic efficacy of CA inhibitors (CAIs) in disease models, bridging the gap between laboratory findings and clinical applications. The therapeutic relevance of CAIs extends across multiple domains, including glaucoma, epilepsy, cancer, metabolic disorders, and infectious diseases. Emerging applications, such as their potential use in combating antimicrobial resistance and modulating immune responses, further underscore their versatility. However, challenges such as achieving isoform selectivity, minimizing off-target effects, and translating preclinical findings into clinical success persist. Advances in fragment-based drug design, artificial intelligence-driven discovery, and innovative experimental techniques are poised to address these limitations, paving the way for the next generation of CAIs.
Collapse
Affiliation(s)
- Ahmed A Allam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Hassan A Rudayni
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Noha A Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Faris F Aba Alkhayl
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Al Mokhtar Lamsabhi
- Departamento de Química and Institute for advanced research in chemical Science (IAdChem), Facultad de Ciencias, Módulo 13, Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Emadeldin M Kamel
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt.
| |
Collapse
|
8
|
Li Y, Yang W, Chen H, Jin Z, Dong J, Ma L, Ji Z. Comprehensive pan-cancer single-cell analysis reveals glycolysis-related signatures as predictive biomarkers for immunotherapy response and their role in bladder cancer. Int Immunopharmacol 2025; 152:114381. [PMID: 40058104 DOI: 10.1016/j.intimp.2025.114381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/24/2025]
Abstract
Glycolysis is a vital metabolic biological process in tumor progression and immune modulation. This study comprehensively investigated the roles of glycolysis in pan-cancer, especially in bladder cancer. Exploration of 34 single-cell RNA sequencing (scRNA-seq) cohorts, eight ICI-treated bulk RNA-seq cohorts, and TCGA bulk pan-cancer RNA-seq cohorts uncovered a Glycolysis.Sig which strongly correlated with immunotherapy response and demonstrated excellent predictive performance in prognosis and immune response. Hub-Glycolysis.Sig exhibited varying interactions with the immune microenvironment based on cancer type. In bladder cancer, higher glycolysis risk scores correlated with poorer prognosis, with distinct immune infiltration characteristics between subtypes. scRNA-seq revealed high glycolysis levels in bladder epithelial cells. COPB2 was highly expressed in bladder cancer, promoting cell proliferation, migration, and glycolytic activity in vitro and in vivo. Our large-scale data analysis confirmed the negative correlation between glycolysis and immunotherapy outcomes, identifying Glycolysis.Sig as a novel predictive biomarker. Hub-Glycolysis.Sig provides clinical insights for bladder cancer therapy strategies, while COPB2 and other potential therapeutic targets facilitate personalized cancer treatment.
Collapse
Affiliation(s)
- Yingjie Li
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Wenjie Yang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Hualin Chen
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Zhaoheng Jin
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Jie Dong
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Lin Ma
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China.
| | - Zhigang Ji
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China.
| |
Collapse
|
9
|
Kim EY, Abides J, Keller CR, Martinez SR, Li W. Tumor Microenvironment Lactate: Is It a Cancer Progression Marker, Immunosuppressant, and Therapeutic Target? Molecules 2025; 30:1763. [PMID: 40333742 PMCID: PMC12029365 DOI: 10.3390/molecules30081763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/12/2025] [Accepted: 04/12/2025] [Indexed: 05/09/2025] Open
Abstract
The "Warburg effect" is a term coined a century ago for the preferential use of glycolysis over aerobic respiration in tumor cells for energy production, even under aerobic conditions. Although this is a less efficient mechanism of generating energy from glucose, aerobic glycolysis, in addition to the canonical anaerobic glycolysis, is an effective means of lactate production. The abundant waste product, lactate, yielded by the dual glycolysis in a tumor, has been discovered to be a major biomolecule that drives cancer progression. Lactate is a metabolic energy source that, via cell membrane lactate transporters, shuttles in and out of cancer cells as well as cancer cell-associated stromal cells and immune cells within the tumor microenvironment (TME). Additionally, lactate serves as a pH tuner, signaling ligand and transducer, epigenetic and gene transcription regulator, TME modifier, immune suppressor, chemoresistance modulator, and prognostic marker. With such broad functionalities, the production-consumption-reproduction of TME lactate fuels tumor growth and dissemination. Here, we elaborate on the lactate sources that contribute to the pool of lactate in the TME, the functions of TME lactate, the influence of the TME lactate on immune cell function and local tissue immunity, and anticancer therapeutic approaches adopting lactate manipulations and their efficacies. By scrutinizing these properties of the TME lactate and others that have been well addressed in the field, it is expected that a better weighing of the influence of the TME lactate on cancer development, progression, prognosis, and therapeutic efficacy can be achieved.
Collapse
Affiliation(s)
- Eugene Y. Kim
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (E.Y.K.); (J.A.); (C.R.K.)
| | - Joyce Abides
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (E.Y.K.); (J.A.); (C.R.K.)
- Doctor of Medicine Program, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| | - Chandler R. Keller
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (E.Y.K.); (J.A.); (C.R.K.)
| | - Steve R. Martinez
- Department of Medical Education and Clinical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
- Providence Regional Cancer Partnership, Providence Regional Medical Center, Everett, WA 98201, USA
| | - Weimin Li
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (E.Y.K.); (J.A.); (C.R.K.)
| |
Collapse
|
10
|
Zhang Q, Song X, Liu J, Zhou X. Prospects of engineered bacteria-assisted CAR T Cell therapy in gastrointestinal cancers. Oncol Rev 2025; 19:1581856. [PMID: 40297102 PMCID: PMC12034723 DOI: 10.3389/or.2025.1581856] [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: 02/23/2025] [Accepted: 04/03/2025] [Indexed: 04/30/2025] Open
Abstract
The high incidence and mortality rates associated with gastrointestinal cancers represent a significant global health challenge. In recent years, CAR T cell therapy has emerged as a promising immunotherapeutic approach, demonstrating favorable clinical outcomes. However, the application of traditional CAR T cell therapy in gastrointestinal cancers faces numerous challenges, including the suppressive tumor microenvironment and limitations in anti-tumor efficacy. The application of engineered bacteria offers a novel strategy to enhance CAR T cell therapy by modulating the tumor microenvironment and boosting immune responses, potentially leading to improved therapeutic outcomes. This review synthesizes the current research advancements related to engineered bacteria-assisted CAR T cell therapy in gastrointestinal cancers, exploring its underlying mechanisms, clinical applications, and future developmental directions.
Collapse
Affiliation(s)
- Qingqing Zhang
- Reproductive Medicine, Weifang People’s Hospital, Weifang, Shandong, China
| | - Xiao Song
- Department of Gastroenterology, Weifang People’s Hospital, Weifang, Shandong, China
| | - Junhong Liu
- The Third Department of Geriatrics, Weifang People’s Hospital, Weifang, Shandong, China
| | - Xuejiao Zhou
- Hospital Preparation Center, Weifang People’s Hospital, Weifang, Shandong, China
| |
Collapse
|
11
|
Tang J, Chen L, Shen X, Xia T, Li Z, Chai X, Huang Y, Yang S, Peng X, Lai J, Li R, Xie L. Exploring the Role of Cellular Interactions in the Colorectal Cancer Microenvironment. J Immunol Res 2025; 2025:4109934. [PMID: 40255905 PMCID: PMC12008489 DOI: 10.1155/jimr/4109934] [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: 08/23/2024] [Accepted: 02/22/2025] [Indexed: 04/22/2025] Open
Abstract
Colorectal cancer (CRC) stands as one of the tumors with globally high incidence and mortality rates. In recent years, researchers have extensively explored the role of the tumor immune microenvironment (TME) in CRC, highlighting the crucial influence of immune cell populations in driving tumor progression and shaping therapeutic outcomes. The TME encompasses an array of cellular and noncellular constituents, spanning tumor cells, immune cells, myeloid cells, and tumor-associated fibroblasts, among others. However, the cellular composition within the TME is highly dynamic, evolving throughout different stages of tumor progression. These shifts in cell subpopulation proportions lead to a gradual transition in the immune response, shifting from an early antitumor growth to a late-stage environment that supports tumor survival. Therefore, it is crucial to further investigate and understand the complex interactions among the various cell populations within the TME. In this review, we explore the key cellular components of varying origins, subpopulations with shared origins, and noncellular elements within the CRC TME, examining their interconnections and critical considerations for developing personalized and precise immunotherapy strategies.
Collapse
Affiliation(s)
- Jiadai Tang
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Liuhan Chen
- Department of Head and Neck Surgery Section II, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Xin Shen
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Tingrong Xia
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Zhengting Li
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Xiaoying Chai
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Yao Huang
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Shaoqiong Yang
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Xinjun Peng
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Junbo Lai
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Rui Li
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Lin Xie
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| |
Collapse
|
12
|
Lu C, Shang J, Xie M, Zhu Y, Zhong J, He Y, Xiao Z, Chen W, Yang ZA, Tang X, Yin P, Chen J. Bufalin inhibits immune escape in metastatic colorectal cancer by regulating M2 macrophage polarization. Apoptosis 2025:10.1007/s10495-025-02107-y. [PMID: 40186793 DOI: 10.1007/s10495-025-02107-y] [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] [Accepted: 03/22/2025] [Indexed: 04/07/2025]
Abstract
The prognosis for patients with metastatic colorectal cancer (mCRC) remains poor primarily owing to immune escape caused by immunosuppressive tumor microenvironment (TME). M2 tumor-associated macrophages (TAMs) have been considered as a pivotal role in sustaining the immunosuppressive character in TME. Our previous studies have found that highly mCRC cells could promote M2 TAMs polarization, leading to the exhaustion of T cell antitumor immunity. Studies have reported that Bufalin (BU) could reverse the immunosuppressive TME via regulating TAMs polarization, but the mechanisms underlying remain elusive. In this study, we demonstrated that KLF4 secreted by highly mCRC cells not only promoted the polarization to M2 TAMs but also up-regulated the PD-L1 expression in TAMs, leading to suppressing cytotoxic T lymphocyte (CTL) function to facilitate tumor immune escape. Mechanistically, BU targeted the SRC-3 protein to reduce KLF4 release in highly mCRC cells to regulate the polarization of M2 TAMs and down-regulate PD-L1 expression in TAMs, resulting in reprogramming of the TME and enhancing the anti-tumor immunity. These results have also been validated in both subcutaneous tumor models and orthotopic tumor models. Overall, this research further elucidates the anti-tumor mechanism of BU for inhibiting immune escape in mCRC and facilitate exploitation of a new potential macrophage-based mCRC immunotherapeutic modality.
Collapse
Affiliation(s)
- Chang Lu
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- Fifth Clinical Medical College, Anhui Medical University, Hefei, 230022, Anhui, China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Jing Shang
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- Department of Radiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Manli Xie
- Department of Medical Oncology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Yuan Zhu
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Jiani Zhong
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Yujie He
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Zengyou Xiao
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- Fifth Clinical Medical College, Anhui Medical University, Hefei, 230022, Anhui, China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Wen Chen
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- Fifth Clinical Medical College, Anhui Medical University, Hefei, 230022, Anhui, China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Ze-An Yang
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
- Fifth Clinical Medical College, Anhui Medical University, Hefei, 230022, Anhui, China
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Xiaoxia Tang
- Department of Medical Oncology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Peihao Yin
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
- Fifth Clinical Medical College, Anhui Medical University, Hefei, 230022, Anhui, China.
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, China.
- Department of General Surgery, Putuo People's Hospital, Tongji University School of Medicine, Shanghai, 200060, China.
- Shanghai Key Laboratory of Wearable Robotics and Human-Machine Interaction, Shanghai, China.
| | - Jinbao Chen
- Department of Medical Oncology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| |
Collapse
|
13
|
Zhang L, Wei G, Wang K, Han X. Effect of compound kushen injection on immune function in patients with colorectal cancer: a systematic review and meta-analysis. Front Pharmacol 2025; 16:1565031. [PMID: 40248100 PMCID: PMC12003389 DOI: 10.3389/fphar.2025.1565031] [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: 01/22/2025] [Accepted: 03/26/2025] [Indexed: 04/19/2025] Open
Abstract
Background Colorectal cancer (CRC) is one of the most common malignant tumors worldwide. Chemotherapy and radiotherapy remain cornerstone treatments; however, they often lead to significant immune suppression and an increased risk of infection. Enhancing immune function in CRC patients is critical for improving clinical outcomes and prognosis. Objective To evaluate the effects of Compound Kushen Injection (CKI) on immune function and its role in mitigating chemotherapy-induced adverse effects in patients with CRC. Methods We retrieved randomized controlled trials (RCTs) evaluating the effects of CKI on immune function in patients with CRC from eight Chinese and English databases, up until 31 December 2024. The Cochrane Handbook was used to assess the quality of the included studies. For the meta-analysis, we utilized Review Manager 5.4.1 software. Sensitivity analysis and publication bias assessment were conducted using Stata 17.0 software. Result A total of 2,663 patients (1,550 males and 1,113 females) from 30 RCTs were included. Compared to conventional chemotherapy (CC), the combination of CKI with CC significantly enhanced immune function, increasing CD3+ levels (MD = 6.15, 95% CI: 4.78 to 7.53, p < 0.00001), CD4+ levels (MD = 8.05, 95% CI: 6.99 to 9.11, p < 0.00001), CD4+/CD8+ levels (MD = 0.36, 95% CI: 0.28 to 0.44, p < 0.00001), NK cell levels (MD = 3.60, 95% CI: 2.85 to 4.34, p < 0.00001), while reducing CD8+ levels (MD = -4.19, 95% CI: -5.11 to -3.27, p < 0.00001). CKI also improved the objective response rate (ORR, RR = 1.50, 95% CI: 1.38 to 1.62, p < 0.00001) and disease control rate (DCR, RR = 1.15, 95% CI: 1.10 to 1.19, p < 0.00001), decreased CEA levels (MD = -1.79, 95% CI: -2.81 to -0.76, p = 0.0007) and CA199 levels (MD = -0.73, 95% CI: -1.35 to -0.12, p = 0.02), and reduced chemotherapy-induced adverse reactions, including nausea, vomiting, hepatic dysfunction, myelosuppression, neurotoxicity, leukopenia, thrombocytopenia, and mouth ulcers. Conclusion Current evidence suggests that the combination of CKI with CC may have beneficial effects on immune function, ORR, DCR, and chemotherapy-induced adverse reactions in CRC patients. However, given the variability in study quality and the absence of disease stage stratification, these findings should be interpreted with caution. Furthermore, the lack of long-term follow-up data limits the understanding of CKI's impact on survival and quality of life. High-quality, large-scale RCTs with extended follow-up are needed to further assess the long-term efficacy, safety, and clinical applicability of CKI in CRC management. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=632516, identifier CRD42025632516.
Collapse
Affiliation(s)
- Lixin Zhang
- Department of Anorectal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Guangyan Wei
- College of Pharmacy, Yanbian University, Yanji, China
| | - Kaiping Wang
- Department of Anorectal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xu Han
- Department of Anorectal Surgery, Chongqing Changshou Traditional Chinese Medicine Hospital, Chongqing, China
| |
Collapse
|
14
|
Liu F, Gu Z, Yi F, Liu X, Zou W, Xu Q, Yuan Y, Chen N, Tang J. Potential of Glycyrrhiza in the prevention of colitis-associated colon cancer. Fitoterapia 2025; 181:106398. [PMID: 39842555 DOI: 10.1016/j.fitote.2025.106398] [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: 10/16/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Glycyrrhiza, a legume native to the Mediterranean region, has a long history of ethnomedicinal use in China. Due to its antiviral, antibacterial, anti-inflammatory, antioxidant, antitumor, anti-ulcer, and hepatoprotective properties, Glycyrrhiza is widely utilized in the treatment of gastrointestinal disorders. THE AIM OF THE REVIEW The specific mechanisms of the main active constituents of glycyrrhiza in the treatment of inflammatory bowel disease, precancerous lesions and colorectal cancer at all stages of the colitis-associated colon cancer "Inflammation-Dysplasia-Cancer" sequence, as well as its pharmacokinetics, toxicology, formulation improvements, and application studies, are reviewed to provide new insights and perspectives on glycyrrhiza as a dietary supplement to treat and prevent colitis-associated colon cancer. MATERIALS AND METHODS Information on Glycyrrhiza was retrieved from electronic databases, including PubMed and Web of Science. RESULTS Glycyrrhiza is a well-established medicinal plant with significant potential for applications in both the food and pharmaceutical industries. Over 400 active constituents have been identified in Glycyrrhiza, including terpenoids, flavonoids, isoflavones, coumarins, and polyphenols. Numerous studies have demonstrated that Glycyrrhiza and its active compounds can inhibit the "Inflammation-Dysplasia-Cancer" progression of colitis-associated colon cancer by mitigating inflammatory bowel disease, reducing the number of intestinal precancerous lesions, and counteracting colorectal cancer. Furthermore, derivatives and nanocarriers are crucial for the effective treatment of colitis-associated colon cancer using Glycyrrhiza and its active constituents. CONCLUSION In conclusion, Glycyrrhiza is a plant with both medicinal and nutritional value, making it a potential food ingredient and dietary supplement for the treatment of colitis-associated colon cancer.
Collapse
Affiliation(s)
- Fang Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China; North Sichuan Medical College, Nanchong, China.
| | - Zhili Gu
- North Sichuan Medical College, Nanchong, China
| | - Feiyang Yi
- North Sichuan Medical College, Nanchong, China
| | - Xue Liu
- North Sichuan Medical College, Nanchong, China
| | - Wenxuan Zou
- North Sichuan Medical College, Nanchong, China
| | - Qingxia Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yun Yuan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Nianzhi Chen
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jianyuan Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
15
|
Sias F, Zoroddu S, Migheli R, Bagella L. Untangling the Role of MYC in Sarcomas and Its Potential as a Promising Therapeutic Target. Int J Mol Sci 2025; 26:1973. [PMID: 40076599 PMCID: PMC11900228 DOI: 10.3390/ijms26051973] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
MYC plays a pivotal role in the biology of various sarcoma subtypes, acting as a key regulator of tumor growth, proliferation, and metabolic reprogramming. This oncogene is frequently dysregulated across different sarcomas, where its expression is closely intertwined with the molecular features unique to each subtype. MYC interacts with critical pathways such as cell cycle regulation, apoptosis, and angiogenesis, amplifying tumor aggressiveness and resistance to standard therapies. Furthermore, MYC influences the tumor microenvironment by modulating cell-extracellular matrix interactions and immune evasion mechanisms, further complicating therapeutic management. Despite its well-established centrality in sarcoma pathogenesis, targeting MYC directly remains challenging due to its "undruggable" protein structure. However, emerging therapeutic strategies, including indirect MYC inhibition via epigenetic modulators, transcriptional machinery disruptors, and metabolic pathway inhibitors, offer new hope for sarcoma treatment. This review underscores the importance of understanding the intricate roles of MYC across sarcoma subtypes to guide the development of effective targeted therapies. Given MYC's central role in tumorigenesis and progression, innovative approaches aiming at MYC inhibition could transform the therapeutic landscape for sarcoma patients, providing a much-needed avenue to overcome therapeutic resistance and improve clinical outcomes.
Collapse
Affiliation(s)
- Fabio Sias
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari, Italy; (F.S.); (S.Z.)
| | - Stefano Zoroddu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari, Italy; (F.S.); (S.Z.)
| | - Rossana Migheli
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy;
| | - Luigi Bagella
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari, Italy; (F.S.); (S.Z.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Centre for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
16
|
Li S, Ye J, Yang K, Xu C, Qin Z, Xue Y, Yu L, Zhou T, Yin Z, Sun B, Xu J. Targeting the AURKB- MAD2L2 Axis Disrupts the DNA Damage Response and Glycolysis to Inhibit Colorectal Cancer Progression. FRONT BIOSCI-LANDMRK 2025; 30:26532. [PMID: 40018943 DOI: 10.31083/fbl26532] [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: 09/30/2024] [Revised: 11/14/2024] [Accepted: 12/31/2024] [Indexed: 03/01/2025]
Abstract
BACKGROUND Dysregulated metabolic pathways, including glycolysis and a compromised DNA damage response (DDR), are linked to the progression of colorectal cancer (CRC). The mitotic arrest deficient-like 2 (MAD2L2) and aurora kinase B (AURKB) genes play roles in cell cycle regulation and the DDR, making them potential targets for CRC therapy. METHODS Differential expression analysis was performed using The Cancer Genome Atlas-Colon Adenocarcinoma (TCGA-COAD) and GSE47074 datasets. A predictive model was established, and gene expression levels were further analyzed. The Gene Expression Profiling Interaction Analysis database and co-immunoprecipitation experiments assessed the correlation between AURKB and MAD2L2. Knockdown experiments in CRC cell lines further investigated the role of AURKB, followed by analyses of cell behavior, oxidative stress, glycolysis, DDR, and interaction with MAD2L2. RESULTS The risk model identified six prognostic genes (BUB1 mitotic checkpoint serine/threonine kinase B (BUB1B), AURKB, aurora kinase A (AURKA), exonuclease 1 (EXO1), topoisomerase II alpha (TOP2A), cyclin A2 (CCNA2)) associated with CRC, which were significantly expressed in tumor samples from the TCGA-COAD and GSE47074 datasets. In vitro assays confirmed that AURKB knockdown inhibited CRC cell behavior, induced G1 cell cycle arrest, and increased oxidative stress and apoptosis. AURKB knockdown also impaired glycolysis, reducing lactate production, glucose uptake, and ATP levels. Overexpression of MAD2L2 partially reversed these effects, restored glycolytic activity, and mitigated the cell cycle arrest and DDR caused by AURKB knockdown. CONCLUSION AURKB regulates CRC progression by modulating glycolysis and DDR pathways. Targeting the AURKB-MAD2L2 axis offers a promising therapeutic strategy for disrupting fundamental metabolic and DNA repair mechanisms in CRC.
Collapse
Affiliation(s)
- Shengjie Li
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| | - Jiayou Ye
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| | - Kaifeng Yang
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| | - Chengfan Xu
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| | - Zhixiang Qin
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| | - Yiyang Xue
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - Lanjian Yu
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| | - Tianyu Zhou
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| | - Ziming Yin
- Department of Chemistry, University of Warwick Coventry, CV4 7AL Coventry, UK
| | - Bin Sun
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, 200438 Shanghai, China
| | - Jun Xu
- Department of Gastroenterology Surgery, Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, 443000 Yichang, Hubei, China
| |
Collapse
|
17
|
Xu L, Shen Y, Zhang C, Shi T, Sheng X. Exploring the Link Between Noncoding RNAs and Glycolysis in Colorectal Cancer. J Cell Mol Med 2025; 29:e70443. [PMID: 39993964 PMCID: PMC11850098 DOI: 10.1111/jcmm.70443] [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: 12/11/2024] [Revised: 01/22/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Glycolysis is implicated in the onset and progression of colorectal cancer (CRC) through its influence on the proliferation, invasiveness, chemoresistance and immune system evasion of neoplasm cells. Increasing evidence has shown that the abnormal expression of noncoding RNAs (ncRNAs), especially microRNAs (miRNAs), long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs), in CRC is closely related to glycolysis. In this review, we present a synthesis of the latest research insights into the modulatory roles and distinct pathways of ncRNAs in the glycolytic process in CRC. This knowledge may pave the way for identifying novel therapeutic targets, as well as novel prognostic and diagnostic biomarkers for CRC.
Collapse
Affiliation(s)
- Liang Xu
- Neonatal Department, Suzhou Ninth People's HospitalSuzhou Ninth Hospital Affiliated to Soochow UniversitySuzhouJiangsuChina
| | - Yu Shen
- Department of General Surgery, Suzhou Ninth People's HospitalSuzhou Ninth Hospital Affiliated to Soochow UniversitySuzhouJiangsuChina
| | - Chuanqiang Zhang
- Department of General SurgeryThe Affiliated Jiangsu Shengze Hospital of Nanjing Medical UniversitySuzhouChina
- Shengze Clinical Medical CollegeKangda College of Nanjing Medical UniversityNanjingChina
| | - Tongguo Shi
- Jiangsu Institute of Clinical ImmunologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xuejuan Sheng
- Health Management Center, Suzhou Ninth People's HospitalSuzhou Ninth Hospital Affiliated to Soochow UniversitySuzhouJiangsuChina
| |
Collapse
|
18
|
Zhu M, Hu Y, Gu Y, Lin X, Jiang X, Gong C, Fang Z. Role of amino acid metabolism in tumor immune microenvironment of colorectal cancer. Am J Cancer Res 2025; 15:233-247. [PMID: 39949925 PMCID: PMC11815375 DOI: 10.62347/zsoo2247] [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: 11/09/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
This review investigates the role of amino acid metabolism in the tumor microenvironment of colorectal cancer (CRC) and explores potential targeted therapeutic strategies. The paper synthesized current research on amino acid metabolism in the colorectal cancer tumor microenvironment, focusing on amino acids such as tryptophan, methionine, glutamine, and arginine. It examined their impact on tumor growth, immune evasion, and patient prognosis, as well as the metabolic reprogramming of tumor cells and complex tumor microenvironment interactions. Aberrant amino acid metabolism was a hallmark of colorectal cancer, influencing tumor proliferation, survival, and invasiveness. Key findings included: Tryptophan metabolism via the kynurenine and serotonin pathways significantly affected immune response and tumor progression in CRC. Methionine influenced T cell function and DNA methylation, playing a critical role in tumor development. Glutamine was extensively used by tumor cells for energy metabolism and supported immune cell function. Arginine metabolism impacted CD8+ T cell functionality and tumor growth. The review also discussed the dual roles of immune cells in the tumor microenvironment and the potential of targeting amino acid metabolic pathways for CRC treatment. In conclusion, amino acid metabolism significantly impacts the colorectal cancer tumor microenvironment and immunity. Understanding these metabolic pathways provides valuable insights into CRC pathogenesis and identifies potential therapeutic targets. Future research should focus on developing treatments that disrupt these metabolic processes to improve patient outcomes in CRC.
Collapse
Affiliation(s)
- Minjing Zhu
- Clinical Laboratory, Sanmen People’s HospitalSanmen 317100, Zhejiang, China
| | - Yanyan Hu
- Clinical Laboratory, Sanmen People’s HospitalSanmen 317100, Zhejiang, China
| | - Yangjia Gu
- Chinese Medicine, Changchun University of Science and TechnologyChangchun 130600, Jilin, China
| | - Xuedan Lin
- Clinical Laboratory, Sanmen People’s HospitalSanmen 317100, Zhejiang, China
| | - Xiang Jiang
- Department of Gastroenterology, Sanmen People’s HospitalSanmen 317100, Zhejiang, China
| | - Chaoju Gong
- Central Laboratory, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical UniversityXuzhou 221000, Jiangsu, China
| | - Zejun Fang
- Central Laboratory, Sanmen People’s HospitalSanmen 317100, Zhejiang, China
| |
Collapse
|
19
|
Bao Y, Li G, Li S, Zhou H, Yang Z, Wang Z, Yan R, Guo C, Jin Y. A novel nanomedicine integrating ferroptosis and photothermal therapy, well-suitable for PD-L1-mediated immune checkpoint blockade. Mater Today Bio 2024; 29:101346. [PMID: 39635320 PMCID: PMC11616610 DOI: 10.1016/j.mtbio.2024.101346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/03/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024] Open
Abstract
Immunotherapy based on immune checkpoint blockade has emerged as a promising treatment strategy; however, the therapeutic efficacy is limited by the immunosuppressive microenvironment. Here, we developed a novel immune-activated nanoparticle (Fc-SS-Fe/Cu) to address the issue of insufficient immune infiltration. Specifically, the structure of Fc-SS-Fe/Cu collapsed in response to the tumor microenvironment, the ferrocene and disulfide bonds and the released Fe/Cu ions can effectively generate ·OH and deplete GSH to increase oxidative stress, thereby inducing ferroptosis. Withal, the positive feedback mechanisms of "laser-triggered mild-temperature photothermal therapy (PTT), PTT accelerated ferroptosis and LPO accumulation, LPO mediated HSPs down-regulated to promote PTT," effectively triggers immunogenic cell death (ICD) in tumor cells, significantly enhancing their immunogenicity. Moreover, the O2-generating ability induced by Fc-SS-Fe/Cu could reverse the hypoxic tumor microenvironment, and importantly, the expression of PD-L1 on tumor cell surfaces could be effectively downregulated by inhibiting the HIF-1α pathways, thereby augmenting the effect of anti-PD-L1 (αPD-L1) therapy. Therefore, this study provides valuable strategies into enhancing PD-L1-mediated ICB therapy.
Collapse
Affiliation(s)
- Yujun Bao
- Key Laboratory of Molecular Cytogenetics and Genetic Breeding of Heilongjiang Province, College of Life Science and Technology, Harbin Normal University, Harbin, 150025, China
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Guanghao Li
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Siqi Li
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Haishui Zhou
- Key Laboratory of Molecular Cytogenetics and Genetic Breeding of Heilongjiang Province, College of Life Science and Technology, Harbin Normal University, Harbin, 150025, China
| | - Ziqing Yang
- School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Zhiqiang Wang
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Rui Yan
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Changhong Guo
- Key Laboratory of Molecular Cytogenetics and Genetic Breeding of Heilongjiang Province, College of Life Science and Technology, Harbin Normal University, Harbin, 150025, China
| | - Yingxue Jin
- Key Laboratory of Molecular Cytogenetics and Genetic Breeding of Heilongjiang Province, College of Life Science and Technology, Harbin Normal University, Harbin, 150025, China
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| |
Collapse
|
20
|
Stepkowski S, Bekbolsynov D, Oenick J, Brar S, Mierzejewska B, Rees MA, Ekwenna O. The Major Role of T Regulatory Cells in the Efficiency of Vaccination in General and Immunocompromised Populations: A Review. Vaccines (Basel) 2024; 12:992. [PMID: 39340024 PMCID: PMC11436018 DOI: 10.3390/vaccines12090992] [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: 07/17/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Since their conception with the smallpox vaccine, vaccines used worldwide have mitigated multiple pandemics, including the recent COVID-19 outbreak. Insightful studies have uncovered the complexities of different functional networks of CD4 T cells (T helper 1 (Th1); Th2, Th17) and CD8 T cells (T cytotoxic; Tc), as well as B cell (BIgM, BIgG, BIgA and BIgE) subsets, during the response to vaccination. Both T and B cell subsets form central, peripheral, and tissue-resident subsets during vaccination. It has also become apparent that each vaccination forms a network of T regulatory subsets, namely CD4+ CD25+ Foxp3+ T regulatory (Treg) cells and interleukin-10 (IL-10)-producing CD4+ Foxp3- T regulatory 1 (Tr1), as well as many others, which shape the quality/quantity of vaccine-specific IgM, IgG, and IgA antibody production. These components are especially critical for immunocompromised patients, such as older individuals and allograft recipients, as their vaccination may be ineffective or less effective. This review focuses on considering how the pre- and post-vaccination Treg/Tr1 levels influence the vaccination efficacy. Experimental and clinical work has revealed that Treg/Tr1 involvement evokes different immune mechanisms in diminishing vaccine-induced cellular/humoral responses. Alternative steps may be considered to improve the vaccination response, such as increasing the dose, changing the delivery route, and/or repeated booster doses of vaccines. Vaccination may be combined with anti-CD25 (IL-2Rα chain) or anti-programmed cell death protein 1 (PD-1) monoclonal antibodies (mAb) to decrease the Tregs and boost the T/B cell immune response. All of these data and strategies for immunizations are presented and discussed, aiming to improve the efficacy of vaccination in humans and especially in immunocompromised and older individuals, as well as organ transplant patients.
Collapse
Affiliation(s)
- Stanislaw Stepkowski
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, OH 43614, USA; (D.B.); (B.M.)
| | - Dulat Bekbolsynov
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, OH 43614, USA; (D.B.); (B.M.)
| | - Jared Oenick
- Neurological Surgery, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA;
| | - Surina Brar
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, OH 43614, USA; (D.B.); (B.M.)
| | - Beata Mierzejewska
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, OH 43614, USA; (D.B.); (B.M.)
| | - Michael A. Rees
- Department of Urology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (M.A.R.); (O.E.)
| | - Obi Ekwenna
- Department of Urology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (M.A.R.); (O.E.)
| |
Collapse
|