1
|
Cong R, Lu C, Li X, Xu Z, Wang Y, Sun S. Tumor organoids in cancer medicine: from model systems to natural compound screening. PHARMACEUTICAL BIOLOGY 2025; 63:89-109. [PMID: 39893515 PMCID: PMC11789228 DOI: 10.1080/13880209.2025.2458149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/04/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025]
Abstract
CONTEXT The advent of tissue engineering and biomedical techniques has significantly advanced the development of three-dimensional (3D) cell culture systems, particularly tumor organoids. These self-assembled 3D cell clusters closely replicate the histopathological, genetic, and phenotypic characteristics of primary tissues, making them invaluable tools in cancer research and drug screening. OBJECTIVE This review addresses the challenges in developing in vitro models that accurately reflect tumor heterogeneity and explores the application of tumor organoids in cancer research, with a specific focus on the screening of natural products for antitumor therapies. METHODS This review synthesizes information from major databases, including Chemical Abstracts, Medicinal and Aromatic Plants Abstracts, ScienceDirect, Google Scholar, Scopus, PubMed and Springer Link. Publications were selected without date restrictions, using terms such as 'organoid', 'natural product', 'pharmacological', 'extract', 'nanomaterial' and 'traditional uses'. Articles related to agriculture, ecology, synthetic work or published in languages other than English were excluded. RESULTS AND CONCLUSIONS The review identifies key challenges related to the efficiency and variability of organoid generation and discusses ongoing efforts to enhance their predictive capabilities in drug screening and personalized medicine. Recent studies utilizing patient-derived organoid models for natural compound screening are highlighted, demonstrating the potential of these models in developing new classes of anticancer agents. The integration of natural products with patient-derived organoid models presents a promising approach for discovering novel anticancer compounds and elucidating their mechanisms of action.
Collapse
Affiliation(s)
- Rong Cong
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Can Lu
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xinying Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yaqin Wang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Shusen Sun
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, USA
| |
Collapse
|
2
|
Chen S, Wang Y, Da Zhou, Hu J. Bayesian Inference of Phenotypic Plasticity of Cancer Cells Based on Dynamic Model for Temporal Cell Proportion Data. Biom J 2025; 67:e70055. [PMID: 40298362 DOI: 10.1002/bimj.70055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/18/2024] [Accepted: 10/31/2024] [Indexed: 04/30/2025]
Abstract
Mounting evidence underscores the prevalent hierarchical organization of cancer tissues. At the foundation of this hierarchy reside cancer stem cells, a subset of cells endowed with the pivotal role of engendering the entire cancer tissue through cell differentiation. In recent times, substantial attention has been directed toward the phenomenon of cancer cell plasticity, where the dynamic interconversion between cancer stem cells and nonstem cancer cells has garnered significant interest. Since the task of detecting cancer cell plasticity from empirical data remains a formidable challenge, we propose a Bayesian statistical framework designed to infer phenotypic plasticity within cancer cells, utilizing temporal data on cancer stem cell proportions. Our approach is grounded in a stochastic model, adept at capturing the dynamic behaviors of cells. Leveraging Bayesian analysis, we scrutinize the moment equation governing cancer stem cell proportions, derived from the Kolmogorov forward equation of our stochastic model. Our methodology introduces an improved Euler method for parameter estimation within nonlinear ordinary differential equation models, also extending insights to compositional data. Extensive simulations robustly validate the efficacy of our proposed method. To further corroborate our findings, we apply our approach to analyze published data from SW620 colon cancer cell lines. Our results harmonize with in situ experiments, thereby reinforcing the utility of our method in discerning and quantifying phenotypic plasticity within cancer cells.
Collapse
Affiliation(s)
- Shuli Chen
- School of Mathematics, Sun Yat-sen University, Guangzhou, Guangdong, China
- School of Mathematical Science, Xiamen University, Xiamen, Fujian, China
| | - Yuman Wang
- School of Mathematical Science, Xiamen University, Xiamen, Fujian, China
| | - Da Zhou
- School of Mathematical Science, Xiamen University, Xiamen, Fujian, China
| | - Jie Hu
- School of Mathematical Science, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
3
|
Li M, Liu B, Xian M, Wang S, Liu P. Bioinformatics combined with network pharmacology and experimental validation to identify key biomarkers of hepatocellular carcinoma and corresponding compounds in Radix Astragali and Pueraria Mirifica. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5351-5371. [PMID: 39549064 DOI: 10.1007/s00210-024-03597-4] [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: 09/30/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024]
Abstract
The occurrence and death rates of primary hepatocellular carcinoma (HCC) are increasing, and there remains a shortage of effective oral medications with minimal side effects. We aim to identify potential biomarkers and compounds from Radix Astragali (RA) and Pueraria Mirifica (PM) to treat liver cancer and improve prognosis. Differentially expressed genes (DEGs) associated with HCC were identified by bioinformatics analysis of three datasets, GSE112791, GSE101685, and GSE45114. Using public databases to predict the bioactive components and possible targets of RA and PM. Target crossover from Gene Expression Omnibus (GEO) and public databases were used to identify potential biomarkers for HCC. Subsequently, validation and prognostic value analyses were performed using the Gene Expression Profile Interaction Analysis (GEPIA) platform. The Cytoscape software created a network of "compound targets" to pinpoint compounds linked to the biomarkers. Molecular docking techniques were utilized to validate the connection between these compounds and the identified biomarkers. Ultimately, the HepG2 liver cancer cell line was chosen to assess the inhibitory effect of Hederagenin (HDG) and to confirm the expression of ADH1B through Western blot analysis. In this study, four key biomarkers (NR1I2, ADH1B, NQO1, GHR) were identified. Molecular docking showed that these four core targets could form stable conformations with the corresponding compounds. As the drug concentration decreases, the inhibitory effect on HepG2 diminishes, and the survival rate of HepG2 cells significantly declines following the administration of 100 µmol/L HDG. Compared to the control, the expression of ADH1B protein is significantly increased in HepG2 cells treated with 100 µmol/L HDG. The study identified four key biomarkers (ADH1B, GHR, NQO1, NR1I2) that have prognostic ability for HCC. This study provides biomarkers and potential targeted monomeric medicines for treating HCC.
Collapse
Affiliation(s)
- Mohan Li
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Traditional Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Engineering and Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| | - Bang Liu
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Traditional Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Engineering and Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| | - Minghua Xian
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Traditional Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Engineering and Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shumei Wang
- Guangdong Pharmaceutical University, Guangzhou, 510006, China.
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, School of Traditional Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China.
- Engineering and Technology Research Center for Chinese Materia Medica Quality of the Universities of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China.
- Traditional Chinese Medicine Resource Germplasm Bank Management Center, Yunfu, 527300, China.
| | - Peiyi Liu
- Huangpu People's Hospital of Zhongshan, Zhongshan, 528429, China
| |
Collapse
|
4
|
Lu M, Zhang T, Yang Y, Lin X, Huang J, Sun Y, Sun T. MXene Loaded With Cu (2- x )Se Nanozyme for Nanocatalytic Tumor Therapy. Chemistry 2025; 31:e202500574. [PMID: 40107883 DOI: 10.1002/chem.202500574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 03/22/2025]
Abstract
Traditional tumor treatments (surgery, radiotherapy, chemotherapy, etc.) have certain limitations and can have serious negative effects, such as difficulty in cutting out tumors, damage to normal tissues, and complications. Ordinary nanozymes have low catalytic activity and require higher doses for treatment, which can increase in vivo toxicity and side effects. To address these limitations, we developed a Ti3C2 MXene-based nanocomposite (Ti3C2/Cu(2- x )Se) integrating Cu(2- x )Se nanozymes with dual enzyme-mimicking activities (catalase and peroxidase) and MXene's photothermal properties. The Cu(2- x )Se component catalyzes the decomposition of tumor-overexpressed H2O2 into O2 and cytotoxic ·OH, alleviating hypoxia while inducing oxidative stress. Simultaneously, MXene's high surface area and photothermal capability enhance nanozyme stability, biocompatibility, and catalytic efficiency under near-infrared irradiation. Notably, the photothermal effect amplifies enzymatic activity, enabling synergistic nanocatalytic-photothermal therapy. This synergy not only degrades glutathione to suppress tumor antioxidant defenses but also achieves targeted tumor ablation with reduced dosage requirements. Our work highlights a rational design of MXene-based nanozymes for enhanced multimodal tumor therapy, offering a paradigm for nanocomposite-driven disease treatment.
Collapse
Affiliation(s)
- Mengtian Lu
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, People's Republic of China
| | - Tianye Zhang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, People's Republic of China
| | - Yue Yang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, People's Republic of China
| | - Xin Lin
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, People's Republic of China
| | - Jin Huang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, People's Republic of China
| | - Yuan Sun
- Center of Pharmaceutical Engineering and Technology, Harbin University of Commerce, Harbin, People's Republic of China
| | - Tiedong Sun
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, People's Republic of China
| |
Collapse
|
5
|
Tapiainen VV, Sirniö P, Elomaa H, Karjalainen H, Äijälä VK, Kastinen M, Kehusmaa A, Pohjanen VM, Lindgren O, Sirkiä O, Ahtiainen M, Helminen O, Wirta EV, Rintala J, Saarnio J, Rautio T, Seppälä TT, Böhm J, Mecklin JP, Tuomisto A, Mäkinen MJ, Väyrynen JP. Stroma AReactive Invasion Front Areas (SARIFA), tumour immune microenvironment, and survival in colorectal cancer. Br J Cancer 2025; 132:805-813. [PMID: 40055484 PMCID: PMC12041369 DOI: 10.1038/s41416-025-02972-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/05/2025] [Accepted: 02/25/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND SARIFA (Stroma AReactive Invasion Front Areas), defined as the direct contact between a tumour cell cluster and adipose cells at the invasion margin, has been proposed as a prognostic marker in gastrointestinal cancers. We hypothesized that SARIFA is associated with an immunosuppressive tumour microenvironment. METHODS SARIFA status was evaluated in two large colorectal cancer cohorts (N = 1876). Survival analyses were performed using multivariable Cox regression. Immune cell densities were analysed utilizing multiplex and conventional immunohistochemistry combined with digital image analysis. RESULTS SARIFA-positivity was independently associated with a shorter cancer-specific survival in both cohorts [Cohort 1: hazard ratio (HR) for SARIFA-positive (vs. negative) 1.75 (95% CI 1.35-2.25), P < 0.0001; Cohort 2: HR for SARIFA-positive (vs. negative) 2.09 (95% CI 1.43-3.05), P = 0.0001]. SARIFA-positivity was associated with lower densities of CD3+ T cells, CD66b+ granulocytes, M1-like macrophages, and CD14+HLA-DR+ mature monocytic cells, but higher densities of M2-like macrophages and CD14+HLA-DR- immature monocytic cells. Mean Cohen's kappa for SARIFA evaluation between eight investigators was 0.80. CONCLUSIONS SARIFA status is a highly reproducible, independent prognostic factor in colorectal cancer. SARIFA-positivity is associated with lower densities of antitumourigenic immune cells and the polarisation of macrophages towards a protumourigenic M2-like phenotype.
Collapse
Affiliation(s)
- Vilja V Tapiainen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Päivi Sirniö
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Hanna Elomaa
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
- Department of Education and Research, Hospital Nova of Central Finland, Well Being Services County of Central Finland, Jyväskylä, Finland
| | - Henna Karjalainen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Ville K Äijälä
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Meeri Kastinen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Akseli Kehusmaa
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Vesa-Matti Pohjanen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Outi Lindgren
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Onni Sirkiä
- Department of Pathology, Hospital Nova of Central Finland, Well Being Services County of Central Finland, Jyväskylä, Finland
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Maarit Ahtiainen
- Central Finland Biobank, Hospital Nova of Central Finland, Well Being Services County of Central Finland, Jyväskylä, Finland
| | - Olli Helminen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Erkki-Ville Wirta
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
| | - Jukka Rintala
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Juha Saarnio
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Tero Rautio
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Toni T Seppälä
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere University Hospital, Tampere, Finland
- Department of Gastrointestinal Surgery, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
- Applied Tumour Genomics, Research Program Unit, University of Helsinki, Helsinki, Finland
| | - Jan Böhm
- Department of Pathology, Hospital Nova of Central Finland, Well Being Services County of Central Finland, Jyväskylä, Finland
| | - Jukka-Pekka Mecklin
- Department of Education and Research, Hospital Nova of Central Finland, Well Being Services County of Central Finland, Jyväskylä, Finland
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Anne Tuomisto
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Markus J Mäkinen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Juha P Väyrynen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland.
| |
Collapse
|
6
|
Qiao K, Huang Y, Ning S, Lyu M, Xie J, Zhang S, Lu X, Yu Y, Jiang W, Liu B, Fan K, Liu T. Camouflaged Nanozymes with Oxidation-Promoting Activities Triggering Ferroptosis for Radio-Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417370. [PMID: 40285563 DOI: 10.1002/advs.202417370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/14/2025] [Indexed: 04/29/2025]
Abstract
Radioresistance presents a substantial obstacle to achieving optimal therapeutic outcomes for breast cancer treatment. In this study, we develop a cancer cell membrane (CM) - coated nanozyme system (MPPC@CM), specifically designed for radioimmunotherapy to address this issue. This innovative system involves the in situ reduction of platinum and palladium on mesoporous silica nanospheres, followed by functionalization with cinnamaldehyde via surface grafting. The CM coating endows the nanozyme with enhanced tumor-specific targeting capability due to its homing properties. Upon uptake by tumor cells, MPPC@CM catalytically generates O2 from H2O2, mitigating the hypoxic tumor microenvironment and reducing radioresistance. The intracellular glutathione depletion mediated by Michael addition reactions concurrently disrupts endogenous antioxidant defenses against reactive oxygen species (ROS). This redox imbalance is synergistically amplified through nanozyme-mediated catalytic activities including both peroxidase-like and oxidase-like functions. The resultant massive ROS accumulation establishes a self-reinforcing oxidative cascade that ultimately induces functional inactivation of glutathione peroxidase 4. The immunosuppressive environment is remodeled by this disturbance in redox balance, which accelerates ferroptosis and increases CD8+ T-cell infiltration and dendritic cell maturation. Overall, this cell membrane-camouflaged nanozyme holds significant potential to enhance the efficacy of radioimmunotherapy.
Collapse
Affiliation(s)
- Kun Qiao
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150000, China
- Key Laboratory of Tumor Biotherapy of Heilongjiang Province, Harbin Medical University Cancer Hospital, Harbin, 150000, China
| | - Yongbiao Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Shipeng Ning
- Research Center of Nanomedicine Technology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Meng Lyu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jieqiong Xie
- Research Center of Nanomedicine Technology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Shiyuan Zhang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150000, China
| | - Xiuxin Lu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150000, China
| | - Yuan Yu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150000, China
| | - Wei Jiang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, 450000, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450000, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450000, China
| | - Tong Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150000, China
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, China
| |
Collapse
|
7
|
Luan X, Peng X, Hou Q, Liu J. LINC00892 as a Prognostic Biomarker in Lung Adenocarcinoma: Role in Immune Infiltration and EMT Suppression. J Immunol Res 2025; 2025:4341348. [PMID: 40308809 PMCID: PMC12041620 DOI: 10.1155/jimr/4341348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 03/10/2025] [Indexed: 05/02/2025] Open
Abstract
Lung adenocarcinoma (LUAD) is a prevalent and aggressive form of lung cancer with poor prognosis, largely due to late-stage diagnosis and limited therapeutic options. Recent studies suggest that long noncoding RNAs (lncRNAs) play critical roles in cancer progression and immune modulation, emerging as potential therapeutic targets. In this study, we investigated the expression and functional role of LINC00892 in LUAD using RNA sequencing data from The Cancer Genome Atlas (TCGA) and functional assays in vitro and in vivo. We found that LINC00892 is significantly downregulated in LUAD tissues compared to normal tissues, and lower LINC00892 expression correlates with poorer overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI), particularly in younger patients and those with early-stage disease. Bioinformatic analyses revealed that LINC00892 expression is positively correlated with immune cell infiltration, including CD4+ and CD8+ T cells, and negatively correlated with tumor-promoting Th2 cells, suggesting its role in shaping the tumor immune microenvironment. In vitro functional assays showed that LINC00892 overexpression inhibits LUAD cell proliferation, migration, and invasion while promoting apoptosis. Mechanistically, LINC00892 upregulation was found to suppress epithelial-mesenchymal transition (EMT) by increasing E-cadherin expression and decreasing levels of N-cadherin, vimentin, and slug. Additionally, in an in vivo mouse xenograft model, LINC00892 overexpression suppressed tumor growth and metastasis, accompanied by enhanced immune cell infiltration such as CD4+ and CD8+ T cells. Collectively, these findings suggest that LINC00892 acts as a tumor suppressor in LUAD by modulating immune infiltration and EMT, highlighting its potential as a prognostic biomarker and therapeutic target.
Collapse
Affiliation(s)
- Xinyu Luan
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Xuxing Peng
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Qinghua Hou
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jixian Liu
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
8
|
Giram P, Md Mahabubur Rahman K, Aqel O, You Y. In Situ Cancer Vaccines: Redefining Immune Activation in the Tumor Microenvironment. ACS Biomater Sci Eng 2025. [PMID: 40223683 DOI: 10.1021/acsbiomaterials.5c00121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Cancer is one of the leading causes of mortality worldwide. Nanomedicines have significantly improved life expectancy and survival rates for cancer patients in current standard care. However, recurrence of cancer due to metastasis remains a significant challenge. Vaccines can provide long-term protection and are ideal for preventing bacterial and viral infections. Cancer vaccines, however, have shown limited therapeutic efficacy and raised safety concerns despite extensive research. Cancer vaccines target and stimulate responses against tumor-specific antigens and have demonstrated great potential for cancer treatment in preclinical studies. However, tumor-associated immunosuppression and immune tolerance driven by immunoediting pose significant challenges for vaccine design. In situ vaccination represents an alternative approach to traditional cancer vaccines. This strategy involves the intratumoral administration of immunostimulants to modulate the growth and differentiation of innate immune cells, such as dendritic cells, macrophages, and neutrophils, and restore T-cell activity. Currently approved in situ vaccines, such as T-VEC, have demonstrated clinical promise, while ongoing clinical trials continue to explore novel strategies for broader efficacy. Despite these advancements, failures in vaccine research highlight the need to address tumor-associated immune suppression and immune escape mechanisms. In situ vaccination strategies combine innate and adaptive immune stimulation, leveraging tumor-associated antigens to activate dendritic cells and cross-prime CD8+ T cells. Various vaccine modalities, such as nucleotide-based vaccines (e.g., RNA and DNA vaccines), peptide-based vaccines, and cell-based vaccines (including dendritic, T-cell, and B-cell approaches), show significant potential. Plant-based viral approaches, including cowpea mosaic virus and Newcastle disease virus, further expand the toolkit for in situ vaccination. Therapeutic modalities such as chemotherapy, radiation, photodynamic therapy, photothermal therapy, and Checkpoint blockade inhibitors contribute to enhanced antigen presentation and immune activation. Adjuvants like CpG-ODN and PRR agonists further enhance immune modulation and vaccine efficacy. The advantages of in situ vaccination include patient specificity, personalization, minimized antigen immune escape, and reduced logistical costs. However, significant barriers such as tumor heterogeneity, immune evasion, and logistical challenges remain. This review explores strategies for developing potent cancer vaccines, examines ongoing clinical trials, evaluates immune stimulation methods, and discusses prospects for advancing in situ cancer vaccination.
Collapse
Affiliation(s)
- Prabhanjan Giram
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Kazi Md Mahabubur Rahman
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Osama Aqel
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Youngjae You
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| |
Collapse
|
9
|
Zhang S, Pan J, Guo H, Guan X, Yan C, Ji L, Wu X, Huangfu H. Prognostic value and immunotherapy analysis of immune cell-related genes in laryngeal cancer. PeerJ 2025; 13:e19239. [PMID: 40247837 PMCID: PMC12005187 DOI: 10.7717/peerj.19239] [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: 10/08/2024] [Accepted: 03/11/2025] [Indexed: 04/19/2025] Open
Abstract
Background Laryngeal cancer (LC) is a prevalent head and neck carcinoma. Extensive research has established a link between immune cells in the tumor microenvironment (TME) and cancer progression, as well as responses to immunotherapy. This study aims to develop a prognostic model based on immune cell-related genes and examine the TME in LC. Methods RNA-seq data for LC were sourced from The Cancer Genome Atlas (TCGA), and GSE27020 and GSE51985 datasets were retrieved from the Gene Expression Omnibus (GEO) database. Key genes were identified through the intersection of differentially expressed genes (DEGs) between normal and LC samples and module genes derived from weighted gene co-expression network analysis (WGCNA), followed by functional enrichment analysis. The prognostic risk model was constructed using univariate Cox and Least Absolute Shrinkage and Selection Operator (LASSO) analyses. Gene Set Variation Analysis (GSVA) was subsequently performed for hallmark and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses in high- and low-risk groups. Immune infiltration analysis between risk groups was conducted via Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) and single sample gene set enrichment analysis (ssGSEA). Finally, the relationship between the risk model and immunotherapy response was explored. Results A total of 124 key genes were identified through the overlap analysis, predominantly enriched in GO terms such as defense response to viruses and regulation of response to biotic stimuli, as well as KEGG pathways related to phagosome and Epstein-Barr virus infection. Machine learning indicated that the optimal prognostic model was constructed from two biomarkers, RENBP and OLR1. GSVA revealed that in the high-risk group, epithelial-mesenchymal transition and ECM-receptor interaction were the most significantly enriched pathways, while autoimmune thyroid disease, ribosome, and oxidative phosphorylation predominated in the low-risk group. Additionally, the stromal score was significantly higher in the high-risk group, while CD8+ T cells, cytolytic activity, inflammation promotion, and T cell co-stimulation were elevated in the low-risk group. Tumor Immune Dysfunction and Exclusion (TIDE) analysis showed higher TIDE and exclusion scores in the high-risk group, whereas the CD8 score was higher in the low-risk group. Finally, CD274 (PD-L1) expression was significantly elevated in the low-risk group. Conclusions This study identified two key prognostic biomarkers, RENBP and OLR1, and characterized TME differences across risk groups, offering novel insights into the diagnosis and treatment of LC.
Collapse
Affiliation(s)
- Sen Zhang
- The First Hospital of Shanxi Medical University, Department of Otolaryngology Head and Neck Surgery, Taiyuan, Shanxi, China
- Shanxi Medical University, First Clinical Medical College, Taiyuan, Shanxi, China
- Shanxi Medical University, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan, Shanxi, China
| | - Jianrui Pan
- The First Hospital of Shanxi Medical University, Department of Otolaryngology Head and Neck Surgery, Taiyuan, Shanxi, China
- Shanxi Medical University, First Clinical Medical College, Taiyuan, Shanxi, China
- Shanxi Medical University, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan, Shanxi, China
| | - Huina Guo
- The First Hospital of Shanxi Medical University, Department of Otolaryngology Head and Neck Surgery, Taiyuan, Shanxi, China
- Shanxi Medical University, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan, Shanxi, China
| | - Xiaoya Guan
- The First Hospital of Shanxi Medical University, Department of Otolaryngology Head and Neck Surgery, Taiyuan, Shanxi, China
- Shanxi Medical University, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan, Shanxi, China
| | - Chenxu Yan
- The First Hospital of Shanxi Medical University, Department of Otolaryngology Head and Neck Surgery, Taiyuan, Shanxi, China
- Shanxi Medical University, First Clinical Medical College, Taiyuan, Shanxi, China
- Shanxi Medical University, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan, Shanxi, China
| | - Lingling Ji
- The First Hospital of Shanxi Medical University, Department of Otolaryngology Head and Neck Surgery, Taiyuan, Shanxi, China
- Shanxi Medical University, First Clinical Medical College, Taiyuan, Shanxi, China
- Shanxi Medical University, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan, Shanxi, China
| | - Xiansha Wu
- The First Hospital of Shanxi Medical University, Department of Otolaryngology Head and Neck Surgery, Taiyuan, Shanxi, China
- Shanxi Medical University, First Clinical Medical College, Taiyuan, Shanxi, China
- Shanxi Medical University, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan, Shanxi, China
| | - Hui Huangfu
- The First Hospital of Shanxi Medical University, Department of Otolaryngology Head and Neck Surgery, Taiyuan, Shanxi, China
- Shanxi Medical University, First Clinical Medical College, Taiyuan, Shanxi, China
- Shanxi Medical University, Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan, Shanxi, China
| |
Collapse
|
10
|
Wan X, Ashaolu TJ, Sun MC, Zhao C. A Review of Food Bioactives That Can Modulate miRNA Profiles for Management of Colorectal Cancer. Foods 2025; 14:1352. [PMID: 40282754 PMCID: PMC12027151 DOI: 10.3390/foods14081352] [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: 03/01/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
Colorectal cancer (CRC), the third leading cause of cancer globally, with high mortality, necessitates more effective treatments and adjunct therapies. MicroRNAs (miRNAs), short non-coding RNAs, regulate gene expression. Food-derived active components have the potential to modulate CRC cellular processes, aiding in the prevention and management of CRC. This review explores the role of miRNAs in CRC and summarizes the anti-inflammatory, antioxidant, and pro-apoptotic effects of typical food bioactive components by modulating specific miRNAs. We investigate the potential and scientific basis of regulating miRNA expression through dietary therapy and preventive approaches, providing new directions for CRC treatment. Collectively, miRNAs regulate gene expression, impacting the onset, progression, metastasis, and treatment response of CRC. Food components such as curcumin and resveratrol modulate specific miRNAs, affecting CRC cell behavior. Bioactive food components influence CRC cell proliferation, apoptosis, and drug sensitivity by regulating key proteins and pathways.
Collapse
Affiliation(s)
- Xiaoqin Wan
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Tolulope Joshua Ashaolu
- Institute for Global Health Innovations, Duy Tan University, Da Nang 550000, Vietnam;
- Faculty of Medicine, Duy Tan University, Da Nang 550000, Vietnam
| | - Mao-Cheng Sun
- College of Food Science and Engineering, Changchun University, Changchun 130022, China
| | - Changhui Zhao
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| |
Collapse
|
11
|
Soleimani Mamalo A, Pashaei M, Valilo M, Ojarudi M. Cytokine-mediated regulation of immune cell metabolic pathways in the tumor microenvironment. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04133-8. [PMID: 40220026 DOI: 10.1007/s00210-025-04133-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Cancer, an important global health problem, is defined by aberrant cell proliferation and continues to be the main cause of death globally. The tumor microenvironment (TME) plays an essential role in the development of cancer, resistance to therapy, and regulation of the immune response. Some immune cells in the TME, like T cells, B cells, macrophages, dendritic cells, and natural killer cells, can either stop or help tumor growth, depending on how metabolic and cytokine changes happen. Cytokines function as essential signaling molecules that modulate immune cell metabolism, altering their functionality. This review focuses on how cytokine-mediated metabolic reprogramming affects the activity of immune cells inside the TME, which can either make the immune response stronger or weaker. New ways of treating cancer that focus on metabolic pathways and cytokine signaling, such as using IL (Interleukin) - 15, IL- 10, and IL- 4, show promise in boosting immune cell activity and making cancer treatments more effective. Finding these pathways could lead to new ways to treat cancer with immunotherapy that focus on metabolic competition and immune resistance in the TME.
Collapse
Affiliation(s)
| | - Mohammadreza Pashaei
- Department of Internal Medicine, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Valilo
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Masoud Ojarudi
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
12
|
Dong H, Ye C, Ye X, Yan J, Ye G, Shao Y. The biological role and molecular mechanism of transfer RNA-derived small RNAs in tumor metastasis. Front Oncol 2025; 15:1560943. [PMID: 40265011 PMCID: PMC12011605 DOI: 10.3389/fonc.2025.1560943] [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: 01/15/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Tumor metastasis is a significant contributor to increased cancer mortality. Transfer RNA-derived small RNAs (tsRNAs), a class of endogenous non-coding RNA molecules, play crucial functional roles in various physiological processes, including the regulation of transcription and reverse transcription, the modulation of translation processes, the modification of epigenetic inheritance, the regulation of the cell cycle, etc. Dysregulated tsRNAs are closely related to the occurrence and progression of human malignancies. Accumulating evidence indicates that the abnormal expression of tsRNAs is associated with tumor metastasis through a variety of mechanisms. Hence, we summarize the fundamental structure and biological functions of tsRNAs, with a focus on how tsRNAs influence the tumor metastasis process through downstream targets or the regulation of interactions between upstream and downstream molecules, thereby providing a novel perspective for targeted therapy for tumor metastasis.
Collapse
Affiliation(s)
- Haotian Dong
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, China
- Health Science Center, Ningbo University, Ningbo, China
| | - Chengyuan Ye
- Health Science Center, Ningbo University, Ningbo, China
| | - Xiaohan Ye
- Health Science Center, Ningbo University, Ningbo, China
| | - Jianing Yan
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Guoliang Ye
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yongfu Shao
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, China
- Health Science Center, Ningbo University, Ningbo, China
| |
Collapse
|
13
|
Chang X, Huang Y, Qu Y, Guo Y, Fan W, Zhen H. Integrative analysis of mitochondrial-related gene profiling identifies prognostic clusters and drug resistance mechanisms in low-grade glioma. Discov Oncol 2025; 16:465. [PMID: 40186003 PMCID: PMC11971116 DOI: 10.1007/s12672-025-02201-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
Mitochondrial dysfunction has emerged as a critical factor in the progression and prognosis of low-grade glioma (LGG). In this study, we explored the role of mitochondrial-related genes through consensus clustering analysis using multi-omics data from the TCGA, CGGA, and other independent datasets. Patients were categorized into three clusters (Cluster A, B, and C), with Cluster B consistently associated with poorer prognosis. Mutation landscape analysis revealed distinct genetic alterations and copy number variations among clusters, particularly in Cluster B, which exhibited unique genetic signatures. Immune infiltration analysis showed that Cluster B had higher expression levels of immune checkpoint genes, stronger immune evasion activity, and greater immune cell infiltration, suggesting an immunosuppressive tumor microenvironment. Furthermore, we identified mitochondrial-related prognostic markers and developed a MITscore based on gene expression patterns, which stratified patients into high- and low-risk groups. High MITscore groups displayed stronger stemness characteristics, poorer survival outcomes, and differential responses to chemotherapy and immunotherapy. Cross-validation with drug sensitivity and immunotherapy cohorts indicated that high MITscore patients were more sensitive to certain chemotherapeutic agents and responded better to immunotherapy. Finally, using the SRGA method, we identified novel biomarkers (KDR, LRRK2, SQSTM1) closely associated with mitochondrial function, which may serve as potential targets for therapeutic intervention. These findings highlight the critical role of mitochondrial dysfunction in LGG prognosis, tumor microenvironment regulation, and treatment response, providing new avenues for precision oncology.
Collapse
Affiliation(s)
- Xiaozan Chang
- Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, Zhengzhou, 450003, Henan, China
| | - Yingxuan Huang
- Pediatric Intensive Care Unit, The Affiliated Hospital of Youjiang Medical University for Nationalities; Key Laboratory of Research and Development on Clinical Molecular Diagnosis for High-Incidence Diseases of Baise, Baise, China
| | - Ying Qu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yu Guo
- Nanfang Hospital (ZengCheng Branch), Southern Medical University, No. 28, Innovation Avenue, Ningxi Street, Guangzhou, China.
| | - Wenwen Fan
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Nanli, Panjiayuan, Beijing, 100021, China.
| | - Haining Zhen
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
14
|
Liu S, Liu C, He Y, Li J. Benign non-immune cells in tumor microenvironment. Front Immunol 2025; 16:1561577. [PMID: 40248695 PMCID: PMC12003390 DOI: 10.3389/fimmu.2025.1561577] [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: 01/20/2025] [Accepted: 02/24/2025] [Indexed: 04/19/2025] Open
Abstract
The tumor microenvironment (TME) is a highly complex and continuous evolving ecosystem, consisting of a diverse array of cellular and non-cellular components. Among these, benign non-immune cells, including cancer-associated fibroblasts (CAFs), adipocytes, endothelial cells (ECs), pericytes (PCs), Schwann cells (SCs) and others, are crucial factors for tumor development. Benign non-immune cells within the TME interact with both tumor cells and immune cells. These interactions contribute to tumor progression through both direct contact and indirect communication. Numerous studies have highlighted the role that benign non-immune cells exert on tumor progression and potential tumor-promoting mechanisms via multiple signaling pathways and factors. However, these benign non-immune cells may play different roles across cancer types. Therefore, it is important to understand the potential roles of benign non-immune cells within the TME based on tumor heterogeneity. A deep understanding allows us to develop novel cancer therapies by targeting these cells. In this review, we will introduce several types of benign non-immune cells that exert on different cancer types according to tumor heterogeneity and their roles in the TME.
Collapse
Affiliation(s)
- Shaowen Liu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chunhui Liu
- The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Yuan He
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jun Li
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
15
|
Pan Z, Liu Y, Li H, Qiu H, Zhang P, Li Z, Wang X, Tian Y, Feng Z, Zhu S, Wang X. The role and mechanism of aerobic glycolysis in nasopharyngeal carcinoma. PeerJ 2025; 13:e19213. [PMID: 40191756 PMCID: PMC11971989 DOI: 10.7717/peerj.19213] [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: 06/20/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
This review delves into the pivotal role and intricate mechanisms of aerobic glycolysis in nasopharyngeal carcinoma (NPC). NPC, a malignancy originating from the nasopharyngeal epithelium, displays distinct geographical and clinical features. The article emphasizes the significance of aerobic glycolysis, a pivotal metabolic alteration in cancer cells, in NPC progression. Key enzymes such as hexokinase 2, lactate dehydrogenase A, phosphofructokinase 1, and pyruvate kinase M2 are discussed for their regulatory functions in NPC glycolysis through signaling pathways like PI3K/Akt and mTOR. Further, the article explores how oncogenic signaling pathways and transcription factors like c-Myc and HIF-1α modulate aerobic glycolysis, thereby affecting NPC's proliferation, invasion, metastasis, angiogenesis, and immune evasion. By elucidating these mechanisms, the review aims to advance research and clinical practice in NPC, informing the development of targeted therapeutic strategies that enhance treatment precision and reduce side effects. Overall, this review offers a broad understanding of the multifaceted role of aerobic glycolysis in NPC and its potential impact on therapeutic outcomes.
Collapse
Affiliation(s)
- Zhiyong Pan
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuyi Liu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Hui Li
- Department of Ophthalmology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Huisi Qiu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Pingmei Zhang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhiying Li
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xinyu Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuxiao Tian
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhengfu Feng
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Song Zhu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xin Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| |
Collapse
|
16
|
Sobolev V, Tchepourina E, Soboleva A, Denisova E, Korsunskaya I, Mezentsev A. PPAR-γ in Melanoma and Immune Cells: Insights into Disease Pathogenesis and Therapeutic Implications. Cells 2025; 14:534. [PMID: 40214488 PMCID: PMC11989151 DOI: 10.3390/cells14070534] [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: 03/06/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Changes in skin pigmentation, like hyperpigmentation or moles, can affect appearance and social life. Unlike locally containable moles, malignant melanomas are aggressive and can spread rapidly, disproportionately affecting younger individuals with a high potential for metastasis. Research has shown that the peroxisome proliferator-activated receptor gamma (PPAR-γ) and its ligands exhibit protective effects against melanoma. As a transcription factor, PPAR-γ is crucial in functions like fatty acid storage and glucose metabolism. Activation of PPAR-γ promotes lipid uptake and enhances sensitivity to insulin. In many cases, it also inhibits the growth of cancer cell lines, like breast, gastric, lung, and prostate cancer. In melanoma, PPAR-γ regulates cell proliferation, differentiation, apoptosis, and survival. During tumorigenesis, it controls metabolic changes and the immunogenicity of stromal cells. PPAR-γ agonists can target hypoxia-induced angiogenesis in tumor therapy, but their effects on tumors can be suppressive or promotional, depending on the tumor environment. Published data show that PPAR-γ-targeting agents can be effective in specific groups of patients, but further studies are needed to understand lesser-known biological effects of PPAR-γ and address the existing safety concerns. This review provides a summary of the current understanding of PPAR-γ and its involvement in melanoma.
Collapse
Affiliation(s)
- Vladimir Sobolev
- Laboratory of Physicochemical and Genetic Problems in Dermatology, Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Moscow 109029, Russia; (V.S.); (E.T.); (A.S.); (E.D.); (I.K.)
| | - Ekaterina Tchepourina
- Laboratory of Physicochemical and Genetic Problems in Dermatology, Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Moscow 109029, Russia; (V.S.); (E.T.); (A.S.); (E.D.); (I.K.)
| | - Anna Soboleva
- Laboratory of Physicochemical and Genetic Problems in Dermatology, Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Moscow 109029, Russia; (V.S.); (E.T.); (A.S.); (E.D.); (I.K.)
| | - Elena Denisova
- Laboratory of Physicochemical and Genetic Problems in Dermatology, Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Moscow 109029, Russia; (V.S.); (E.T.); (A.S.); (E.D.); (I.K.)
- Moscow Center of Dermatovenerology and Cosmetology, Moscow 119071, Russia
| | - Irina Korsunskaya
- Laboratory of Physicochemical and Genetic Problems in Dermatology, Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Moscow 109029, Russia; (V.S.); (E.T.); (A.S.); (E.D.); (I.K.)
| | - Alexandre Mezentsev
- Laboratory of Physicochemical and Genetic Problems in Dermatology, Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Moscow 109029, Russia; (V.S.); (E.T.); (A.S.); (E.D.); (I.K.)
| |
Collapse
|
17
|
Mandloi S, Gargano SM, Duffy AN, Benedict PA, Shing SR, Kahn C, Hannikainen P, Pineda-Reyes JP, Bray D, Toskala EM, Rabinowitz M, Rosen M, Farrell C, Evans JJ, Nyquist GG. The Presence of Pigment Incontinence in Sinonasal Mucosal Melanoma. Laryngoscope 2025; 135:1321-1325. [PMID: 39543905 DOI: 10.1002/lary.31901] [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: 07/29/2024] [Revised: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Regression is an immunological phenomenon described in cutaneous melanoma whereby tumor is replaced with tumor-infiltrating lymphocytes, granulation tissue, and mature fibroblasts often accompanied by pigment incontinence (accumulation of melanin in the upper dermis). Pigment incontinence results in grossly pigmented lesions that may be mistaken for viable tumor and has not been described in sinonasal mucosal melanoma (SNMM). This study investigates the presence of regression and pigment incontinence in patients with SNMM. METHODS A retrospective chart review was conducted on SNMM patients from 2007 to 2023. Pathology slides from surgical resection were examined by two pathologists blinded to treatment information for the presence and extent of pigment-laden macrophages and other histopathologic features of regression. RESULTS Seventeen patients with SNMM were included in this study who underwent surgical resection. Three patients received neoadjuvant therapy followed by surgical resection. Regression was present in 94% of patients and pigment incontinence was present in 65% of patients and occurred in both neoadjuvant treated patients and treatment naïve patients. All three patients with neoadjuvant treatment had evidence of pigment incontinence. DISCUSSION This study highlights that SNMM often displays characteristics of regression. This study is one of the first to describe the presence of pigment incontinence in patients with SNMM. Pigment incontinence can be a part of the natural tumor life cycle and grossly pigmented lesions could easily be confused for melanoma especially after neoadjuvant therapy. Developing an understanding of regression and pigment incontinence within SNMM is important for diagnosis and clinical management. LEVEL OF EVIDENCE 4 Laryngoscope, 135:1321-1325, 2025.
Collapse
Affiliation(s)
- Shreya Mandloi
- Department of Otolaryngology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | | | - Alexander N Duffy
- Department of Otolaryngology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Peter A Benedict
- Department of Otolaryngology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Samuel R Shing
- Department of Otolaryngology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Chase Kahn
- Department of Otolaryngology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Paavali Hannikainen
- Department of Otolaryngology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Juan Pablo Pineda-Reyes
- Department of Pathology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - David Bray
- Department of Neurosurgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Elina M Toskala
- Department of Otolaryngology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
- Department of Neurosurgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Mindy Rabinowitz
- Department of Otolaryngology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
- Department of Neurosurgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Marc Rosen
- Department of Otolaryngology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
- Department of Neurosurgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Christopher Farrell
- Department of Otolaryngology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
- Department of Neurosurgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - James J Evans
- Department of Otolaryngology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
- Department of Neurosurgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| | - Gurston G Nyquist
- Department of Otolaryngology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
- Department of Neurosurgery, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| |
Collapse
|
18
|
Guo Z, Li K, Ren X, Wang X, Yang D, Ma S, Zeng X, Zhang P. The role of the tumor microenvironment in HNSCC resistance and targeted therapy. Front Immunol 2025; 16:1554835. [PMID: 40236700 PMCID: PMC11996806 DOI: 10.3389/fimmu.2025.1554835] [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: 01/03/2025] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
The prognosis for head and neck squamous cell carcinoma (HNSCC) remains unfavorable, primarily due to significant therapeutic resistance and the absence effective interventions. A major obstacle in cancer treatment is the persistent resistance of cancer cells to a variety of therapeutic modalities. The tumor microenvironment (TME) which includes encompasses all non-malignant components and their metabolites within the tumor tissue, plays a crucial role in this context. The distinct characteristics of the HNSCC TME facilitate tumor growth, invasion, metastasis, and resistance to treatment. This review provides a comprehensive overview of the HNSCC TME components, with a particular focus on tumor-associated macrophages (TAMs), regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs), the extracellular matrix, reprogrammed metabolic processes, and metabolic products. It elucidates their contributions to modulating resistance to chemotherapy, radiotherapy, targeted therapy, and immunotherapy in HNSCC, and explores novel therapeutic strategies targeting the TME for HNSCC management.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Peng Zhang
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| |
Collapse
|
19
|
Lugovoi ME, Karshieva SS, Usatova VS, Voznyuk AA, Zakharova VA, Levin AA, Petrov SV, Senatov FS, Mironov VA, Belousov VV, Koudan EV. The design of the spheroids-based in vitro tumor model determines its biomimetic properties. BIOMATERIALS ADVANCES 2025; 169:214178. [PMID: 39799900 DOI: 10.1016/j.bioadv.2025.214178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 12/25/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Cancer, one of the world's deadliest diseases, is expected to claim an estimated 16 million lives by 2040. Three-dimensional (3D) models of cancer have become invaluable tools for the study of tumor biology and the development of new therapies. The tumor microenvironment (TME) is a determinant of tumor progression and has implications for clinical therapies. Cancer-associated fibroblasts (CAFs) are one of the most important components of the TME. Modeling the interactions between cancer cells and CAFs in vitro can help to create biomimetic tumor equivalents for elucidating the causes of cancer growth and assessing the effectiveness of therapies. Here, we are investigated the effect of the mutual arrangement of tumor cells and fibroblasts on the formation of tumor models and their biomimetic properties. Pancreatic tumor models of three different designs were formed by the bioprinting method. Gelatin-alginate hydrogels with and without PANC-1 (pancreatic cancer) and NIH/3 T3 (mouse fibroblasts) cells, as well as their homo- and heterospheroids, were used as bioink. To enable bioprinting, we have chosen the most suitable compositions of alginate and gelatin that provide both good printability and cell proliferation activity. We also have investigated the kinetics of spheroid formation to identify the optimal cultivation parameters for achieving spheroid sizes suitable for bioprinting. All tumor models remained viable for 3-4 weeks. At the same time, the patterns of model development in the cultivation process and the biomimetic properties of the final tissue-engineered structures depended on the model design.
Collapse
Affiliation(s)
- Maksim E Lugovoi
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Saida Sh Karshieva
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Veronika S Usatova
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117997 Moscow, Russia
| | - Amina A Voznyuk
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Vasilina A Zakharova
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Aleksandr A Levin
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Stanislav V Petrov
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Fedor S Senatov
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Vladimir A Mironov
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Vsevolod V Belousov
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117997 Moscow, Russia
| | - Elizaveta V Koudan
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia.
| |
Collapse
|
20
|
Liu J, Liu J, Wang Y, Chen F, He Y, Xie X, Zhong Y, Yang C. Bioactive mesoporous silica materials-assisted cancer immunotherapy. Biomaterials 2025; 315:122919. [PMID: 39481339 DOI: 10.1016/j.biomaterials.2024.122919] [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: 07/14/2024] [Revised: 10/12/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
Immunotherapy is initially envisioned as a powerful approach to train immune cells within the tumor microenvironment (TME) and lymphoid tissues to elicit strong anti-tumor responses. However, clinical cancer immunotherapy still faces challenges, such as limited immunogenicity and insufficient immune response. Leveraging the advantages of mesoporous silica (MS) materials in controllable drug and immunomodulator release, recent efforts have focused on engineering MS with intrinsic immunoregulatory functions to promote robust, systemic, and safe anti-tumor responses. This review discusses advances in bioactive MS materials that address the challenges of immunotherapy. Beyond their role in on-demand delivery and drug release in response to the TME, we highlight the intrinsic functions of bioactive MS in orchestrating localized immune responses by inducing immunogenic cell death in tumor cells, modulating immune cell activity, and facilitating tumor-immune cell interactions. Additionally, we emphasize the advantages of bioactive MS in recruiting and activating immune cells within lymphoid tissues to initiate anti-tumor vaccination. The review also covers the challenges of MS-assisted immunotherapy, potential solutions, and future outlooks. With a deeper understanding of material-bio interactions, the rational design of MS with sophisticated bioactivities and controllable responsiveness holds great promise for enhancing the outcomes of personalized immunotherapy.
Collapse
Affiliation(s)
- Jiali Liu
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, China
| | - Jiying Liu
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yaxin Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, China
| | - Fangman Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Yan He
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Xiaochun Xie
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, 510006, China
| | - Yiling Zhong
- College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 511443, China.
| | - Chao Yang
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China.
| |
Collapse
|
21
|
Karimi S, Bakhshali R, Bolandi S, Zahed Z, Mojtaba Zadeh SS, Kaveh Zenjanab M, Jahanban Esfahlan R. For and against tumor microenvironment: Nanoparticle-based strategies for active cancer therapy. Mater Today Bio 2025; 31:101626. [PMID: 40124335 PMCID: PMC11926801 DOI: 10.1016/j.mtbio.2025.101626] [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: 12/09/2024] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Cancer treatment is challenged by the tumor microenvironment (TME), which promotes drug resistance and cancer cell growth. This review offers a comprehensive and innovative perspective on how nanomedicine can modify the TME to enhance therapy. Strategies include using nanoparticles to improve oxygenation, adjust acidity, and alter the extracellular matrix, making treatments more effective. Additionally, nanoparticles can enhance immune responses by activating immune cells and reducing suppression within tumors. By integrating these approaches with existing therapies, such as chemotherapy and radiotherapy, nanoparticles show promise in overcoming traditional treatment barriers. The review discusses how changes in the TME can enhance the effectiveness of nanomedicine itself, creating a reciprocal relationship that boosts overall efficacy. We also highlight novel strategies aimed at exploiting and overcoming the TME, leveraging nanoparticle-based approaches for targeted cancer therapy through precise TME modulation.
Collapse
Affiliation(s)
- Soroush Karimi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | | | - Zahra Zahed
- Department of Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Masoumeh Kaveh Zenjanab
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Jahanban Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
22
|
Ravi K, Zhang Y, Sakala L, Manoharan TJM, Pockaj B, LaBaer J, Park JG, Nikkhah M. Tumor Microenvironment On-A-Chip and Single-Cell Analysis Reveal Synergistic Stromal-Immune Crosstalk on Breast Cancer Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413457. [PMID: 40056038 PMCID: PMC12021108 DOI: 10.1002/advs.202413457] [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: 10/22/2024] [Revised: 02/11/2025] [Indexed: 04/26/2025]
Abstract
Solid tumors develop within a complex environment called the tumor microenvironment (TME), which is sculpted by the presence of other cells, such as cancer-associated fibroblasts (CAFs) and immune cells like macrophages (Mφs). Despite the presence of immune cells, tumor cells orchestrate a tumor-supportive environment through intricate interaction with the components of the TME. However, the specific mechanism by which this intercellular dialogue is regulated is not fully understood. To that end, the development of an organotypic 3D breast TME-on-a-chip (TMEC) model, integrated with single-cell RNA sequencing analysis, is reported to mechanistically evaluate the progression of triple-negative breast cancer (TNBC) cells in the presence of patient-derived CAFs and Mφs. Extensive functional assays, including invasion and morphometric characterization, reveal the synergistic influence of CAFs and Mφs on tumor cells. Furthermore, gene expression and pathway enrichment analyses identify the involvement of the KYNU gene, suggesting a potential immune evasion mechanism through the kynurenine pathway. Lastly, the pharmacological targeting of the identified pathway is investigated.
Collapse
Affiliation(s)
- Kalpana Ravi
- School of Biological and Health Systems Engineering (SBHSE)Arizona State UniversityTempeAZ85287USA
| | - Yining Zhang
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| | - Lydia Sakala
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| | | | | | - Joshua LaBaer
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| | - Jin G. Park
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE)Arizona State UniversityTempeAZ85287USA
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| |
Collapse
|
23
|
El Saftawy E, Aboulhoda BE, Alghamdi MA, Abd Elkhalek MA, AlHariry NS. Heterogeneity of modulatory immune microenvironment in bladder cancer. Tissue Cell 2025; 93:102679. [PMID: 39700733 DOI: 10.1016/j.tice.2024.102679] [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: 09/23/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Urinary bladder cancer (UBC) is the ninth most common cancer worldwide. The intra-tumor heterogeneity of the UBC microenvironment explains the variances in response to therapy among patients. Tumor immune microenvironment (TIME) is based on the balance between anti-tumor and pro-tumorigenic immunity that eventually determines the tumor fate. This review addresses the recent insights of the cytokines, immune checkpoints, receptors, enzymes, proteins, RNAs, cancer stem cells (CSCs), tissue-resident cells, growth factors, epithelial-mesenchymal transition, microbiological cofactor, and paracrine action of cancer cells that mutually cross-talk within the TIME. In-depth balance and alteration of these factors influence the TIME and the overall tumor progression. This, in turn, highlights the prospects of the new era of manipulating these co-factors for improving the diagnosis, prognosis, and treatment of UBC. CONCLUSION: The heterogenic architecture of the TIME orchestrates the fate of the tumor. Nevertheless, recognizing the mutual cross-talk between these key players seems useful in prognostic and therapeutic approaches.
Collapse
Affiliation(s)
- Enas El Saftawy
- Department of Medical Parasitology, Faculty of Medicine, Cairo University, Cairo, Egypt; Department of Medical Parasitology, Armed Forces College of Medicine, Cairo, Egypt.
| | - Basma Emad Aboulhoda
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Mansour A Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia; Genomics and Personalized Medicine Unit, The Centre for Medical and Heath Research, King Khalid University, Abha 62529, Saudi Arabia
| | - Marwa Ali Abd Elkhalek
- Department of Medical Biochemistry& Molecular Biology, Armed Forces College of Medicine, Cairo, Egypt; Medical Biochemistry & Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | |
Collapse
|
24
|
Chen Y, Zhang Z, Ji K, Zhang Q, Qian L, Yang C. Role of microplastics in the tumor microenvironment (Review). Oncol Lett 2025; 29:193. [PMID: 40041410 PMCID: PMC11877014 DOI: 10.3892/ol.2025.14939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/07/2025] [Indexed: 03/06/2025] Open
Abstract
Microplastics (MPs) are pervasive in several ecosystems and have the potential to infiltrate multiple aspects of human life through ingestion, inhalation and dermal exposure, thus eliciting substantial concerns regarding their potential implications for human health. Whilst initial research has documented the effects of MPs on disease development across multiple physiological systems, MPs may also facilitate tumor progression by influencing the tumor microenvironment (TME). This evolving focus underscores the growing interest in the role of MPs in tumorigenesis and their interactions within the TME. In the present review, the relationship between MPs and the TME is comprehensively assessed, providing a detailed analysis of their interactions with tumor cells, stromal cells (including macrophages, fibroblasts and endothelial cells), the extracellular matrix and inflammatory processes. Recommendations for future research directions and strategies to address and reduce microplastic pollution are proposed.
Collapse
Affiliation(s)
- Yunjie Chen
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zihang Zhang
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Kangming Ji
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qiuchen Zhang
- Department of Radiology, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Lijun Qian
- Department of Geriatric Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Chuang Yang
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
- Department of Radiology, The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| |
Collapse
|
25
|
Shang A, Yu P, Li L, He G, Xu J. Tumor‑stroma ratio as a clinical prognostic factor in colorectal carcinoma: A meta‑analysis of 7,934 patients. Oncol Lett 2025; 29:190. [PMID: 40041409 PMCID: PMC11877013 DOI: 10.3892/ol.2025.14936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/29/2025] [Indexed: 03/06/2025] Open
Abstract
The tumor-stroma ratio (TSR) has been regarded as an important factor associated with tumor metastasis, based on the 'seed and soil' theory, which may have guiding significance for the selection of chemotherapy regimens. Therefore, a high TSR may be a new risk factor for tumor recurrence in patients with stage II colorectal cancer (CRC). The present study aimed to evaluate the prognostic value of TSR in CRC, especially for the computer-calculated TSR. A comprehensive literature retrieval was performed using the PubMed, Web of Science, Embase and Cochrane Library databases to identify relevant studies published up to December 13, 2023. Pooled hazard ratios (HRs) with 95% confidence intervals (CIs) were calculated to estimate the prognostic value of the TSR in CRC. A total of 21 studies published between 2007 and 2023 were included in the present meta-analysis. The combined analysis demonstrated that a high TSR was significantly associated with worse overall survival (OS; HR=1.84; 95% CI, 1.44-2.34; P<0.001), disease-free survival (DFS; HR=1.85; 95% CI, 1.27-2.68; P<0.001), cancer-specific survival (CSS; H=1.97; 95% CI, 1.46-2.65; P<0.001) and recurrence free survival (RFS; HR=1.55; 95% CI, 1.25-1.92; P<0.001) in patients with CRC. Moreover, an elevated computer-calculated TSR was also associated with poor OS (HR=1.89; 95% CI, 1.48-2.40; P<0.001) and DFS (HR=1.85; 95% CI, 1.27-2.68; P<0.001). However, a high TSR was not associated with poor OS in patients with stage I CRC (HR=1.01; 95% CI, 0.48-2.14; P=0.97). In conclusion, the results of the present meta-analysis indicate that a high TSR is associated with poor OS, DFS, CSS and RFS in patients with CRC, especially for those with stage II-III. In addition, TSR calculated by computer using whole-slide images may also be an effective prognostic marker for OS and DFS in patients with CRC.
Collapse
Affiliation(s)
- An Shang
- Department of General Surgery, The Fourth Hospital of Guangxi Medical University, Liuzhou, Guangxi 545007, P.R. China
| | - Pengcheng Yu
- Department of General Surgery, The Fourth Hospital of Guangxi Medical University, Liuzhou, Guangxi 545007, P.R. China
| | - Liping Li
- Department of Pneumology, The Fourth Hospital of Guangxi Medical University, Liuzhou, Guangxi 545007, P.R. China
| | - Ge He
- Department of General Surgery, The Fourth Hospital of Guangxi Medical University, Liuzhou, Guangxi 545007, P.R. China
| | - Junyi Xu
- Department of General Surgery, The Fourth Hospital of Guangxi Medical University, Liuzhou, Guangxi 545007, P.R. China
| |
Collapse
|
26
|
Yuan Z, Li J, Na Q. Recent advances in biomimetic nanodelivery systems for the treatment of glioblastoma. Colloids Surf B Biointerfaces 2025; 252:114668. [PMID: 40168694 DOI: 10.1016/j.colsurfb.2025.114668] [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: 03/04/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Glioblastoma remain one of the deadliest malignant tumors in the central nervous system, largely due to their aggressiveness, high degree of heterogeneity, and the protective barrier of the blood-brain barrier (BBB). Conventional therapies including surgery, chemotherapy and radiotherapy often fail to improve patient prognosis due to limited drug penetration and non-specific toxicity. We then present recent advances in biomimetic nanodelivery systems, focusing on cell membrane coatings, nanoenzymes, and exosome-based carriers. By mimicking endogenous biological functions, these systems demonstrate improved immune evasion, enhanced BBB traversal, and selective drug release within the tumor microenvironment. Nevertheless, we acknowledge unresolved bottlenecks related to large-scale production, stability, and the intricacies of regulatory compliance. Looking forward, we propose an interdisciplinary roadmap that combines materials engineering, cellular biology, and clinical expertise. Through this collaborative approach, this work aims to optimize biomimetic nanodelivery for glioma therapy and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Zhenru Yuan
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Jing Li
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Qi Na
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
27
|
Yang S, Xu X, Wang M, Ma Q, Feng C, Wang J, Zhang Y, Song Y. Gel-to-Coacervate Transition in Peptide/HA Complexes for MMP-9-Activated Penetration into Tumor Spheroids. ACS APPLIED MATERIALS & INTERFACES 2025; 17:18108-18115. [PMID: 40088140 DOI: 10.1021/acsami.4c23089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
Short phase-separating peptides serve as liquid-based vehicles due to their remarkable fluidity and cell permeability, holding great promise in diffusion-limited applications such as intracellular drug delivery or penetration into deep-seated tumors. However, tuning the phase stability and the phase-transition sensitivity of these coacervates in response to specific pathological signals remains a significant challenge. To tackle this challenge, this study presents a phase-separating peptide/hyaluronic acid (HA) complex coacervate system, which undergoes a solid-to-coacervate transition upon exposure to matrix metalloproteinase 9 (MMP-9). By harnessing this disease-relevant enzyme, overexpressed in the ovarian tumor microenvironment, we further demonstrate the improved infiltration of the coacervates into Hey cells and tumor spheroids. These observations highlight the feasibility of modulating phase behaviors and advanced functions of coacervates through sequence-specific monomer design, offering a practical strategy for the on-target delivery of coacervates and medicine into tumors.
Collapse
Affiliation(s)
- Shi Yang
- State Key Laboratory of Metal Matrix Composites, School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiuli Xu
- State Key Laboratory of Metal Matrix Composites, School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Meixin Wang
- State Key Laboratory of Metal Matrix Composites, School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qingming Ma
- School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Chuanliang Feng
- State Key Laboratory of Metal Matrix Composites, School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianwei Wang
- Shanghai Key Laboratory of Embryo Original Diseases and Department of Anesthesiology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yage Zhang
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Yang Song
- State Key Laboratory of Metal Matrix Composites, School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
28
|
Wen L, Wang P, Zhang G, Ma Y, Li J, Chen D, Liu L, Hu H, Huang C, Yao X. Prognostic and Immunological Significance of NMNAT1 in Colorectal and Pan-Cancer Contexts. Onco Targets Ther 2025; 18:389-410. [PMID: 40160196 PMCID: PMC11954486 DOI: 10.2147/ott.s504668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Nicotinamide plays a critical role in the prevention and treatment of tumors, and its metabolism is closely associated with tumor progression. The aim of this study was to understand the prognostic and immunological significance of nicotinamide metabolism-related genes in pan-cancer. Methods We downloaded The Cancer Genome Atlas and Genotype Tissue Expression pan-cancer datasets for NMNAT1 from the UCSC database. We analyzed the differential expression, prognosis, genetic alterations, DNA methylation, immune infiltration, and co-expression with RNA modification-related genes and immune checkpoint-related genes. Genes with expression patterns similar to NMNAT1 were identified using the GEPIA library. The GSCA database was used to investigate the correlation between gene expression and drug sensitivity, as assessed by GDSC and CTRP. The CancerSEA database was employed to examine the association of NMNAT1 expression at the single-cell level across different tumors and its relation to 14 functional states. Immunohistochemistry was performed to assess the clinical significance of NMNAT1 expression. Results NMNAT1 exhibited differential expression across 25 tumor types, including colorectal cancer (CRC), and its expression was significantly associated with the prognosis of 11 tumors. Furthermore, NMNAT1 expression correlated significantly with clinicopathological features. NMNAT1 was strongly associated with immune cells, RNA modification-related genes, and immune checkpoint-related genes in most tumors, affecting immune responses. The expression of NMNAT1 also correlated with sensitivity and resistance to several drugs. Single-cell analysis revealed that NMNAT1 is involved in the progression of retinoblastoma, uveal melanoma, and CRC. Immunohistochemical analysis confirmed that NMNAT1 expression is an independent prognostic factor in patients with CRC. Conclusion NMNAT1 is a crucial prognostic and immune marker gene for nicotinamide metabolism, particularly in CRC. It has potential as a clinical biomarker and a therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Liang Wen
- Gannan Medical University, Ganzhou, People’s Republic of China
- Ganzhou Hospital of Guangdong Provincial People’s Hospital, Ganzhou Municipal Hospital, Ganzhou, People’s Republic of China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
| | - Ping Wang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
| | - Guosheng Zhang
- Ganzhou Hospital of Guangdong Provincial People’s Hospital, Ganzhou Municipal Hospital, Ganzhou, People’s Republic of China
| | - Yongli Ma
- Ganzhou Hospital of Guangdong Provincial People’s Hospital, Ganzhou Municipal Hospital, Ganzhou, People’s Republic of China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
| | - Jinghui Li
- Gannan Medical University, Ganzhou, People’s Republic of China
- Ganzhou Hospital of Guangdong Provincial People’s Hospital, Ganzhou Municipal Hospital, Ganzhou, People’s Republic of China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
| | - Dengzhuo Chen
- Gannan Medical University, Ganzhou, People’s Republic of China
- Ganzhou Hospital of Guangdong Provincial People’s Hospital, Ganzhou Municipal Hospital, Ganzhou, People’s Republic of China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
| | - Linfeng Liu
- Gannan Medical University, Ganzhou, People’s Republic of China
- Ganzhou Hospital of Guangdong Provincial People’s Hospital, Ganzhou Municipal Hospital, Ganzhou, People’s Republic of China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
| | - Hongkai Hu
- Ganzhou Hospital of Guangdong Provincial People’s Hospital, Ganzhou Municipal Hospital, Ganzhou, People’s Republic of China
| | - Chengzhi Huang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
| | - Xueqing Yao
- Gannan Medical University, Ganzhou, People’s Republic of China
- Ganzhou Hospital of Guangdong Provincial People’s Hospital, Ganzhou Municipal Hospital, Ganzhou, People’s Republic of China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
| |
Collapse
|
29
|
Alenezi A, Alhamad H, Alenezi A, Khan MU. Hypoxia Imaging in Lung Cancer: A PET-Based Narrative Review for Clinicians and Researchers. Pharmaceuticals (Basel) 2025; 18:459. [PMID: 40283896 PMCID: PMC12030053 DOI: 10.3390/ph18040459] [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: 03/01/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Hypoxia plays a critical role in lung cancer progression and treatment resistance by contributing to aggressive tumor behavior and poor therapeutic response. Molecular imaging, particularly positron emission tomography (PET), has become an essential tool for noninvasive hypoxia detection, providing valuable insights into tumor biology and aiding in personalized treatment strategies. Objective: This narrative review explores recent advancements in PET imaging for detecting hypoxia in lung cancer, with a focus on the development, characteristics, and clinical applications of various radiotracers. Findings: Numerous PET-based hypoxia radiotracers have been investigated, each with distinct pharmacokinetics and imaging capabilities. Established tracers such as 18F-Fluoromisonidazole (18F-FMISO) remain widely used, while newer alternatives like 18F-Fluoroazomycin Arabinoside (18F-FAZA) and 18F-Flortanidazole (18F-HX4) demonstrate improved clearance and image contrast. Additionally, 64Cu-ATSM has gained attention for its rapid tumor uptake and hypoxia selectivity. The integration of PET with hybrid imaging modalities, such as PET/CT and PET/MRI, enhances the spatial resolution and functional interpretation, making hypoxia imaging a promising approach for guiding radiotherapy, chemotherapy, and targeted therapies. Conclusions: PET imaging of hypoxia offers significant potential in lung cancer diagnosis, treatment planning, and therapeutic response assessment. However, challenges remain, including tracer specificity, quantification variability, and standardization of imaging protocols. Future research should focus on developing next-generation radiotracers with enhanced specificity, optimizing imaging methodologies, and leveraging multimodal approaches to improve clinical utility and patient outcomes.
Collapse
Affiliation(s)
- Ahmad Alenezi
- Radiologic Sciences Department, Kuwait University, Kuwait City 31470, Kuwait
| | - Hamad Alhamad
- Occupational Therapy Department, Kuwait University, Jabriya 31470, Kuwait
| | - Aishah Alenezi
- Radiologic Sciences Department, Kuwait University, Kuwait City 31470, Kuwait
| | - Muhammad Umar Khan
- Nuclear Medicine Department, Jahra Hospital, Ministry of Health, Al Jahra 03200, Kuwait
| |
Collapse
|
30
|
Zhang C, Song Y, Yang H, Wu K. Myeloid cells are involved in tumor immunity, metastasis and metabolism in tumor microenvironment. Cell Biol Toxicol 2025; 41:62. [PMID: 40131539 PMCID: PMC11937113 DOI: 10.1007/s10565-025-10012-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
Bone marrow-derived cells in the tumor microenvironment, including macrophages, neutrophils, dendritic cells, myeloid-derived suppressor cells, eosinophils and basophils, participate in the generation, development, invasion and metastasis of tumors by producing different cytokines and interacting with other cell types, and play a pro-tumor or anti-tumor role in regulating tumor immunity. Due to the complexity of cell types in the tumor microenvironment and the unknown process of tumor development and metastasis, cancer treatment to achieve better survival status remains challenging. In this article, we summarize the effects of myeloid cells in tumor microenvironment on tumor immunity, cancer migration, and crosstalk with metabolism (including glucose metabolism, lipid metabolism, and amino acid metabolism), which will help to further study the tumor microenvironment and seek targeted therapeutic strategies for patients.
Collapse
Affiliation(s)
- Chenbo Zhang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310000, China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310000, China
| | - Ying Song
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310000, China
| | - Huanming Yang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310000, China.
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310000, China.
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Kui Wu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310000, China.
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310000, China.
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
- BGI Genomics, Harbin, 150023, Heilongjiang, China.
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, 518083, China.
| |
Collapse
|
31
|
Dandeu LNR, Lachovsky J, Sidlik S, Marenco P, Orschanski D, Aguilera P, Vázquez M, Carballo MDP, Fernández E, Penas-Steinhardt A, Chasseing NA, Labovsky V. Relevance of oncobiome in breast cancer evolution in an Argentine cohort. mSphere 2025; 10:e0059724. [PMID: 39927763 PMCID: PMC11934308 DOI: 10.1128/msphere.00597-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 01/12/2025] [Indexed: 02/11/2025] Open
Abstract
Breast cancer is the leading cause of cancer deaths in women worldwide, with about 20,000 cases annually in Argentina. While age, diet, and genetics are known risk factors, most breast cancer cases have unknown causes, necessitating the discovery of new risk factors. The aim of this study was the analysis of the prognostic relevance of the oncobiome in Argentinean breast cancer patients. Sequencing of the V4 region 16S rRNA gene was performed on 34 primary breast tumor samples, using bioinformatic and statistical analyses to identify bacteria and hypothetical pathways. Each sample presented a unique microbial profile, with Proteobacteria being the most abundant phylum. Tumors >2 cm showed greater alpha diversity with increased nucleotide biosynthesis. Moreover, progesterone-receptor tumors showed differences in beta diversity, being progesterone receptor-positive tumors that had the highest expression of Acinetobacter and Moraxella. In disease progression, the phylum Chloroflexi was prevalent in tumors of surviving patients. Acinetobacter and Cloacibacterium genera were significantly higher in patients without events and those without metastasis. We found that nucleotide and cell-structure biosynthesis, and lipid metabolism pathways were enriched in tumors with poor progression, whereas amino-acid degradation was increased in tumors of surviving patients. This finding is an indication that tumor cells are taking advantage of this effect of the microbiome during tumor progression. We conclude that oncobiome is dysbiotic in these patients, with distinct patterns in those with poor progression. Suggesting a link between the oncobiome and cancer progression, paving the way for new therapies to improve patient quality of life and survival. IMPORTANCE This is the first study to investigate the relevance of the oncobiome in the evolution of breast cancer in a cohort of Argentine patients. It also highlights the need for further research in this area to improve our understanding of the role of the microbiome in this disease and potentially identify new therapeutic targets or prognostic indicators. Understanding the complex interaction between the microbiome, the tumor microenvironment, and the pathogenesis of breast cancer holds the promise of more personalized and effective treatment approaches in the future.
Collapse
Affiliation(s)
- Leonardo Néstor Rubén Dandeu
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Joel Lachovsky
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Sofía Sidlik
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Marenco
- Instituto de Oncología Ángel H Roffo, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | | | - Pablo Aguilera
- Departamento de Ciencias Aplicadas y Tecnología, Universidad Nacional de Moreno, Moreno, Argentina
| | | | | | - Elmer Fernández
- Fundación para el Progreso de la Medicina, Córdoba, Argentina
| | - Alberto Penas-Steinhardt
- Departamento de Ciencias Básicas, Laboratorio de Genómica Computacional (GEC-UNLu), Universidad Nacional de Luján, Luján, Argentina
| | - Norma Alejandra Chasseing
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Vivian Labovsky
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
32
|
Xiao Q, Tan M, Yan G, Peng L. Revolutionizing lung cancer treatment: harnessing exosomes as early diagnostic biomarkers, therapeutics and nano-delivery platforms. J Nanobiotechnology 2025; 23:232. [PMID: 40119368 PMCID: PMC11929271 DOI: 10.1186/s12951-025-03306-0] [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: 09/28/2024] [Accepted: 03/08/2025] [Indexed: 03/24/2025] Open
Abstract
Lung cancer, known for its high morbidity and mortality rates, remains one of the most critical health challenges globally. Conventional treatment options, such as chemotherapy and surgery, are often limited by high costs, significant side effects, and often yield a poor prognosis. Notably, recent research has shed light on the potential therapeutic roles of exosomes, which essentially influence lung cancer's development, diagnosis, treatment, and prognosis. Exosomes have been revealed for their exceptional properties, including natural intercellular communication, excellent biocompatibility, minimal toxicity, prolonged blood circulation ability, and biodegradability. These unique characteristics position exosomes as highly effective drug delivery systems, nanotherapeutics, and potential diagnostic and prognostic biomarkers in lung cancer. This review provides a comprehensive review of the physiological and pathological roles of exosomes in lung cancer, emphasizing their potential as innovative diagnostic biomarkers, therapeutics, and delivery platforms. By harnessing their unique properties, exosomes are poised to revolutionize the diagnosis and treatment of lung cancer, offering a promising avenue for more personalized and effective therapies.
Collapse
Affiliation(s)
- Qiyao Xiao
- College of Pharmaceutical Sciences, Zhejiang University, 866# Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Minhong Tan
- College of Pharmaceutical Sciences, Zhejiang University, 866# Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Ge Yan
- College of Pharmaceutical Sciences, Zhejiang University, 866# Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Lihua Peng
- College of Pharmaceutical Sciences, Zhejiang University, 866# Yuhangtang Road, Hangzhou, 310058, People's Republic of China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, People's Republic of China.
| |
Collapse
|
33
|
Xin L, Ning S, Wang H, Shi R. Tumor Microenvironment Responsive and Platelet Membrane Coated Polydopamine Nanoparticles for Cancer Radiosensitization by Inducing Cuproptosis. Int J Nanomedicine 2025; 20:3643-3652. [PMID: 40125433 PMCID: PMC11930256 DOI: 10.2147/ijn.s504148] [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: 11/24/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
Background Cuproptosis, distinguished from apoptosis, necroptosis, pyroptosis, and ferroptosis, is a current form of programmed cell death that provides novel strategies for tumor therapy. Nanotechnology inducing cuproptosis showed potential in tumor ablation. However, these strategies might induce cellular damage due to a lack of tumor-targeting ability or insufficient tumor inhibition alone. Methods Here, biomimetic copper-doped polydopamine nanoparticles (PC NPs) were developed to specifically induce tumor cell cuproptosis to enhance radiotherapy (RT). PC NPs were characterized before application for tumor ablation. Results These PC NPs improve tumor targeting and accumulation. After entering the tumor region, PC degrades in cells responsive to acidic tumor microenvironment (TME). Next, Cu2+ is reduced to Cu+ after consuming overexpressed glutathione (GSH), which induces dihydrolipoamide S-acetyltransferase (DLAT) aggression and cuproptosis. Under RT, reactive oxygen species (ROS) are generated and consume GSH, leading to cuproptosis. The decreasing of GSH content in tumor tissues can improve the treatment effect of RT by inhibiting self-repair of tumor cells, hindering cell survival and proliferation. The combination of PC and RT alleviate tumor growth, reaching a tumor growth inhibition rate of 93.0%. Conclusion This tumor-specific targeting nano platform is a valuable radiosensitizer responsive to TME for improving therapeutic efficacy against tumors.
Collapse
Affiliation(s)
- Le Xin
- Department of General Surgery, Longgang Central Hospital of Shenzhen, Shenzhen, 518116, People’s Republic of China
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530031, People’s Republic of China
| | - Hongwei Wang
- Department of General Surgery, Longgang Central Hospital of Shenzhen, Shenzhen, 518116, People’s Republic of China
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
| | - Runze Shi
- The Second Ward of Breast Surgery, Cancer Hospital Affiliated to Harbin Medical University, Harbin, 150086, People’s Republic of China
| |
Collapse
|
34
|
Czajkowski M, Wierzbicki PM, Dolny M, Matuszewski M, Hakenberg OW. Inflammation in Penile Squamous Cell Carcinoma: A Comprehensive Review. Int J Mol Sci 2025; 26:2785. [PMID: 40141426 PMCID: PMC11943298 DOI: 10.3390/ijms26062785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Inflammation appears to play a crucial role in the development and progression of penile cancer (PeCa). Two molecular pathways of PeCa are currently described: HPV-dependent and HPV-independent. The tumor immune microenvironment (TIME) of PeCa is characterized by the presence of tumor-associated macrophages, cancer-associated fibroblasts, and tumor-infiltrating lymphocytes. The components of the TIME produce pro-inflammatory cytokines and chemokines, which have been found to be overexpressed in PeCa tissues and are associated with tumor progression and unfavorable prognoses. Additionally, the nuclear factor kappa B (NF-κB) pathway and secreted phosphoprotein 1 (SPP1) have been implicated in PeCa pathogenesis. Elevated C-reactive protein (CRP) levels and the neutrophil-to-lymphocyte ratio (NLR) have been identified as potential prognostic biomarkers in PeCa. This overview presents the complex contribution of the inflammatory process and collates projects aimed at modulating TIME in PeCa.
Collapse
Affiliation(s)
- Mateusz Czajkowski
- Department of Urology, Medical University of Gdańsk, Mariana Smoluchowskiego 17 Street, 80-214 Gdansk, Poland; (M.D.); (M.M.)
| | - Piotr M. Wierzbicki
- Department of Histology, Medical University of Gdańsk, Dębinki, 80-211 Gdansk, Poland;
| | - Maciej Dolny
- Department of Urology, Medical University of Gdańsk, Mariana Smoluchowskiego 17 Street, 80-214 Gdansk, Poland; (M.D.); (M.M.)
| | - Marcin Matuszewski
- Department of Urology, Medical University of Gdańsk, Mariana Smoluchowskiego 17 Street, 80-214 Gdansk, Poland; (M.D.); (M.M.)
| | - Oliver W. Hakenberg
- Department of Urology, University Medical Center Rostock, 18055 Rostock, Germany;
- Department of Urology, Jena University Hospital, 07747 Jena, Germany
| |
Collapse
|
35
|
Shah DD, Chorawala MR, Raghani NR, Patel R, Fareed M, Kashid VA, Prajapati BG. Tumor microenvironment: recent advances in understanding and its role in modulating cancer therapies. Med Oncol 2025; 42:117. [PMID: 40102282 DOI: 10.1007/s12032-025-02641-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
Tumor microenvironment (TME) denotes the non-cancerous cells and components presented in the tumor, including molecules produced and released by them. Interactions between cancer cells, immune cells, stromal cells, and the extracellular matrix within the TME create a dynamic ecosystem that can either promote or hinder tumor growth and spread. The TME plays a pivotal role in either promoting or inhibiting tumor growth and dissemination, making it a critical factor to consider in the development of effective cancer therapies. Understanding the intricate interplay within the TME is crucial for devising effective cancer therapies. Combination therapies involving inhibitors of immune checkpoint blockade (ICB), and/or chemotherapy now offer new approaches for cancer therapy. However, it remains uncertain how to best utilize these strategies in the context of the complex tumor microenvironment. Oncogene-driven changes in tumor cell metabolism can impact the TME to limit immune responses and present barriers to cancer therapy. Cellular and acellular components in tumor microenvironment can reprogram tumor initiation, growth, invasion, metastasis, and response to therapies. Components in the TME can reprogram tumor behavior and influence responses to treatments, facilitating immune evasion, nutrient deprivation, and therapeutic resistance. Moreover, the TME can influence angiogenesis, promoting the formation of blood vessels that sustain tumor growth. Notably, the TME facilitates immune evasion, establishes a nutrient-deprived milieu, and induces therapeutic resistance, hindering treatment efficacy. A paradigm shift from a cancer-centric model to a TME-centric one has revolutionized cancer research and treatment. However, effectively targeting specific cells or pathways within the TME remains a challenge, as the complexity of the TME poses hurdles in designing precise and effective therapies. This review highlights challenges in targeting the tumor microenvironment to achieve therapeutic efficacy; explore new approaches and technologies to better decipher the tumor microenvironment; and discuss strategies to intervene in the tumor microenvironment and maximize therapeutic benefits.
Collapse
Affiliation(s)
- Disha D Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, Gujarat, 380009, India.
| | - Neha R Raghani
- Department of Pharmacology and Pharmacy Practice, Saraswati Institute of Pharmaceutical Sciences, Gandhinagar, Gujarat, 382355, India
| | - Rajanikant Patel
- Department of Product Development, Granules Pharmaceuticals Inc., 3701 Concorde Parkway, Chantilly, VA, 20151, USA
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, 13713, Riyadh, Saudi Arabia
| | - Vivekanand A Kashid
- MABD Institute of Pharmaceutical Education and Research, Babhulgaon, Yeola, Nashik, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Kherva, Mehsana, Gujarat, 384012, India.
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
36
|
Li J, Liu Z, Zhang G, Yin X, Yuan X, Xie W, Ding X. Uncovering the heterogeneity of NK cells on the prognosis of HCC by integrating bulk and single-cell RNA-seq data. Front Oncol 2025; 15:1570647. [PMID: 40171266 PMCID: PMC11959017 DOI: 10.3389/fonc.2025.1570647] [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: 02/04/2025] [Accepted: 03/04/2025] [Indexed: 04/03/2025] Open
Abstract
Background The tumor microenvironment (TME) plays a critical role in the development, progression, and clinical outcomes of hepatocellular carcinoma (HCC). Despite the critical role of natural killer (NK) cells in tumor immunity, there is limited research on their status within the tumor microenvironment of HCC. In this study, single-cell RNA sequencing (scRNA-seq) analysis of HCC datasets was performed to identify potential biomarkers and investigate the involvement of natural killer (NK) cells in the TME. Methods Single-cell RNA sequencing (scRNA-seq) data were extracted from the GSE149614 dataset and processed for quality control using the "Seurat" package. HCC subtypes from the TCGA dataset were classified through consensus clustering based on differentially expressed genes (DEGs). Weighted gene co-expression network analysis (WGCNA) was employed to construct co-expression networks. Furthermore, univariate and multivariate Cox regression analyses were conducted to identify variables linked to overall survival. The single-sample gene set enrichment analysis (ssGSEA) was used to analyze immune cells and the screened genes. Result A total of 715 DEGs from GSE149614 and 864 DEGs from TCGA were identified, with 25 overlapping DEGs found between the two datasets. A prognostic risk score model based on two genes was then established. Significant differences in immune cell infiltration were observed between high-risk and low-risk groups. Immunohistochemistry showed that HRG expression was decreased in HCC compared to normal tissues, whereas TUBA1B expression was elevated in HCC. Conclusion Our study identified a two-gene prognostic signature based on NK cell markers and highlighted their role in the TME, which may offer novel insights in immunotherapy strategies. Additionally, we developed an accurate and reliable prognostic model, combining clinical factors to aid clinicians in decision-making.
Collapse
Affiliation(s)
- Jiashuo Li
- National Center for Infectious Diseases, Beijing Di’tan Hospital, Capital Medical University, Beijing, China
| | - Zhenyi Liu
- Department of Interventional Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Gongming Zhang
- Department of General Surgery, Beijing You’an Hospital, Capital Medical University, Beijing, China
| | - Xue Yin
- Cancer Center, Beijing Di’tan Hospital, Capital Medical University, Beijing, China
| | - Xiaoxue Yuan
- National Center for Infectious Diseases, Beijing Di’tan Hospital, Capital Medical University, Beijing, China
| | - Wen Xie
- National Center for Infectious Diseases, Beijing Di’tan Hospital, Capital Medical University, Beijing, China
| | - Xiaoyan Ding
- Cancer Center, Beijing Di’tan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
37
|
Liu Z, Chang S, Chen S, Gu R, Guo S. A novel prognostic signature based on m5C‑related LncRNAs and its immunological characteristics in colon adenocarcinoma. Discov Oncol 2025; 16:332. [PMID: 40095128 PMCID: PMC11914420 DOI: 10.1007/s12672-025-02081-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Colon adenocarcinoma (COAD) has high mortality rates due to frequent resistance to treatment. 5-methylcytosine (m5C) is a crucial epigenetic modification of RNA, closely associated with tumorigenesis in various cancers. This study focuses on developing an m5C-related long non-coding RNA (lncRNA) signature to predict prognosis and explore potential therapeutic targets. METHODS Using data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO), we analyzed 18 m5C regulatory genes and their associated lncRNAs in COAD samples. Prognostic lncRNAs were identified through univariate Cox regression, and a risk model was constructed through LASSO regression analyses. Kaplan-Meier survival and receiver operating characteristic analyses were employed to validate the prognostic ability of the signature. Additionally, functional enrichment and immune infiltration analyses were conducted to investigate underlying biological pathways and immune characteristics of the risk groups. Tumor mutation burden and drug sensitivity analyses were also performed. Functional validation of NR2F2-AS1 was conducted through in vitro experiments. RESULTS We established a risk score signature comprising six lncRNAs associated with m5C regulators. Patients were classified into high- and low-risk groups based on the median risk score. This prognostic signature demonstrated significant accuracy and was independent of other clinical features. Immune cell infiltration analysis revealed correlations between the risk signature and various immune cell subtypes. Drug sensitivity analysis indicated the potential therapeutic value of our prognostic signature. Functional experiments confirmed that NR2F2-AS1 acts as a risk factor in the proliferation of colon cancer cells. CONCLUSIONS The m5C-related lncRNA signature serves as a reliable prognostic indicator for colon adenocarcinoma and provides new insights into the tumor immune microenvironment.
Collapse
Affiliation(s)
- Zihe Liu
- Department of Gastroenterology, Jiangsu Funing People's Hospital, Funing, China
| | - Sheng Chang
- Department of Gastroenterology, Jiangsu Funing People's Hospital, Funing, China
| | - Shouguo Chen
- Department of Gastroenterology, Jiangsu Funing People's Hospital, Funing, China
| | - Rong Gu
- Department of Gastroenterology, Jiangsu Funing People's Hospital, Funing, China
| | - Shaoyong Guo
- Department of Gastroenterology, Jiangsu Funing People's Hospital, Funing, China.
| |
Collapse
|
38
|
Che J, Liu Y, Liu Y, Song J, Cui H, Feng D, Tian A, Zhang Z, Xu Y. The application of emerging immunotherapy in the treatment of prostate cancer: progress, dilemma and promise. Front Immunol 2025; 16:1544882. [PMID: 40145100 PMCID: PMC11937122 DOI: 10.3389/fimmu.2025.1544882] [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: 12/13/2024] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
In recent years, there has been a growing trend towards the utilization of immunotherapy techniques for the treatment of cancer. Some malignancies have acquired significant progress with the use of cancer vaccines, immune checkpoint inhibitors, and adoptive cells therapy. Scholars are exploring the aforementioned methods as potential treatments for advanced prostate cancer (PCa) due to the absence of effective adjuvant therapy to improve the prognosis of metastatic castration-resistant prostate cancer (mCRPC). Immunotherapy strategies have yet to achieve significant advancements in the treatment of PCa, largely attributed to the inhibitory tumor microenvironment and low mutation load characteristic of this malignancy. Hence, researchers endeavor to address these challenges by optimizing the design and efficacy of immunotherapy approaches, as well as integrating them with other therapeutic modalities. To date, studies have also shown potential clinical benefits. This comprehensive review analyzed the utilization of immunotherapy techniques in the treatment of PCa, assessing their advantages and obstacles, with the aim of providing healthcare professionals and scholars with a comprehensive understanding of the progress in this field.
Collapse
Affiliation(s)
- Jizhong Che
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yuanyuan Liu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yangyang Liu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Jingheng Song
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Hongguo Cui
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Dongdong Feng
- Department of Urology, Haiyang City People’s Hospital, Yantai, Shandong, China
| | - Aimin Tian
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Zhengchao Zhang
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yankai Xu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
39
|
Tang J, Yang J, Yin LK. Prognostic value of disulfidptosis-associated genes in gastric cancer: a comprehensive analysis. Front Oncol 2025; 15:1512394. [PMID: 40104507 PMCID: PMC11913695 DOI: 10.3389/fonc.2025.1512394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Objective Disulfidptosis is a newly identified type of nonapoptotic programmed cell death related to mechanisms such as ferroptosis, cuproptosis, pyroptosis, and necrotic apoptosis. This study explores the role of disulfidptosis-related long non-coding RNAs (DRLs) in gastric cancer and their potential as prognostic biomarkers. Method We developed a prognostic model using DRL scores to classify patients based on disulfidptosis activity. We evaluated these scores for correlations with drug sensitivity, tumor microenvironment (TME) features, tumor mutational burden (TMB), and prognosis. Potential disulfidptosis-related signaling pathways were screened, identifying FRMD6-AS as a promising therapeutic target. FRMD6-AS expression was further validated using real-time fluorescent quantitative PCR (qRT-PCR). Results The DRL-based prognostic model, established through univariate and multivariate Cox regression and LASSO regression analyses, outperformed traditional models in predicting prognosis. We divided samples into high-risk and low-risk groups based on DRL scores, finding that the low-risk group had a significantly higher survival rate (P < 0.05). A high-precision prediction model incorporating DRL scores, age, sex, grade, and stage showed strong predictive value and consistency with actual outcomes. High DRL scores correlated with higher TME scores and lower TMB. Key signaling axes identified were AC129507.1/(FLNA, TLN1)/FOCAL ADHESION and AC107021.2/MYH10/(TIGHT JUNCTION, VIRAL MYOCARDITIS, REGULATION OF ACTIN CYTOSKELETON). Potentially effective drugs, including BMS-754807, dabrafenib, and JQ1, were identified. FRMD6-AS emerged as a potential target for gastric cancer treatment. Conclusions This study developed a novel prognostic model for gastric cancer using DRLs, identifying two key signaling axes related to prognosis. JQ1 may be an effective treatment, and FRMD6-AS could be a promising therapeutic target.
Collapse
Affiliation(s)
- Jin Tang
- Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jing Yang
- Department of Rheumatology and Immunology, Nanchong Central Hospital, Beijing Anzhen Hospital affiliated to Capital Medical University, Nanchong, Sichuan, China
| | - Long-Kuan Yin
- Department of Gastrointestinal Surgery, People's Hospital of Fushun County, Zigong, Sichuan, China
| |
Collapse
|
40
|
Li M, Wu L, Si H, Wu Y, Liu Y, Zeng Y, Shen B. Engineered mitochondria in diseases: mechanisms, strategies, and applications. Signal Transduct Target Ther 2025; 10:71. [PMID: 40025039 PMCID: PMC11873319 DOI: 10.1038/s41392-024-02081-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/30/2024] [Accepted: 11/17/2024] [Indexed: 03/04/2025] Open
Abstract
Mitochondrial diseases represent one of the most prevalent and debilitating categories of hereditary disorders, characterized by significant genetic, biological, and clinical heterogeneity, which has driven the development of the field of engineered mitochondria. With the growing recognition of the pathogenic role of damaged mitochondria in aging, oxidative disorders, inflammatory diseases, and cancer, the application of engineered mitochondria has expanded to those non-hereditary contexts (sometimes referred to as mitochondria-related diseases). Due to their unique non-eukaryotic origins and endosymbiotic relationship, mitochondria are considered highly suitable for gene editing and intercellular transplantation, and remarkable progress has been achieved in two promising therapeutic strategies-mitochondrial gene editing and artificial mitochondrial transfer (collectively referred to as engineered mitochondria in this review) over the past two decades. Here, we provide a comprehensive review of the mechanisms and recent advancements in the development of engineered mitochondria for therapeutic applications, alongside a concise summary of potential clinical implications and supporting evidence from preclinical and clinical studies. Additionally, an emerging and potentially feasible approach involves ex vivo mitochondrial editing, followed by selection and transplantation, which holds the potential to overcome limitations such as reduced in vivo operability and the introduction of allogeneic mitochondrial heterogeneity, thereby broadening the applicability of engineered mitochondria.
Collapse
Affiliation(s)
- Mingyang Li
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Limin Wu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Haibo Si
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuangang Wu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuan Liu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yi Zeng
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Bin Shen
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
41
|
Khurana S, Sharma S, Goyal PK. Tumor microenvironment as a target for developing anticancer hydrogels. Drug Dev Ind Pharm 2025; 51:157-168. [PMID: 39829011 DOI: 10.1080/03639045.2025.2455424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/28/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE It has been reported that cancer cells get protected by a complex and rich multicellular environment i.e. the tumor microenvironment (TME) consisting of varying immune cells, endothelial cells, dendritic cells, fibroblasts, etc. This manuscript is aimed at the characteristic features of TME considered as potential target(s) for developing smart anticancer hydrogels. SIGNIFICANCE The stimuli-specific drug delivery systems especially hydrogels that can respond to the characteristic features of TME are fabricated for treating cancer. For developing anticancer formulations, TME targeting can be considered an alternative way as it enhances the cytotoxic potential and reduces the unwanted effects. This manuscript shall be of quite interest to academicians, researchers, and clinicians engaged in oncology. METHODS The manuscript was prepared by using the data available in the public domain in online resources such as Google Scholar, PubMed, Science Direct, Scopus, Web of Science, Research Gate, etc. RESULTS Smart hydrogels, sensitive to some specific features of TME such as low pH, high concentration of glutathione, specific enzymes, etc., are promising anticancer formulations as these improve the efficacy and lower the side effects of chemotherapy. CONCLUSION The stimuli-responsive hydrogels have been gaining more attention for delivering cytotoxic drugs to the TME in response to specific stimuli. The stimuli-responsive hydrogels, comprising of cytotoxic drug(s) and specific polymers have some special features such as similarity with biological matrix, ability to respond to various internal as well as external stimuli, improved permeability, porosity, biocompatibility, resemblance with soft living tissues, etc.; and are considered as the promising anticancer candidates.
Collapse
Affiliation(s)
- Suman Khurana
- Amity Institute of Pharmacy, Amity University Haryana, Gurugram, India
- Department of Pharmacy, Panipat Institute of Engineering and Technology, Panipat, India
| | - Shrestha Sharma
- Amity Institute of Pharmacy, Amity University Haryana, Gurugram, India
| | - Parveen Kumar Goyal
- Department of Pharmacy, Panipat Institute of Engineering and Technology, Panipat, India
| |
Collapse
|
42
|
Yang J, Xing Y, Luan J, Yang W, Zhang X, Tian Y, Zhang H. Unveiling tumor-infiltrating immune cell-driven immune-mediated drug resistance in clear cell renal cell carcinoma: prognostic insights and therapeutic strategies. Discov Oncol 2025; 16:259. [PMID: 40025304 PMCID: PMC11872949 DOI: 10.1007/s12672-025-01890-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/03/2025] [Indexed: 03/04/2025] Open
Abstract
INTRODUCTION Tumor drug resistance, particularly immune-mediated resistance, poses a significant challenge in cancer therapy, especially in clear-cell renal cell carcinoma (ccRCC), the most common and aggressive subtype of renal cancer. Tumor-infiltrating immune cells (TIICs) within the tumor microenvironment (TME) play pivotal roles in tumor progression, immune evasion, and therapy resistance. This study explores the prognostic and therapeutic implications of TIICs in ccRCC, aiming to uncover molecular underpinnings and potential strategies to counter drug resistance. METHODS Integrative analyses of transcriptomic and single-cell RNA sequencing data from multiple cohorts were employed to characterize immune and metabolic landscapes in ccRCC. Machine learning algorithms were utilized to identify key TIIC-related RNAs (TIIC-RNAs) associated with prognosis and therapeutic response. The constructed prognostic model was validated across independent datasets. Additionally, the correlation between TIIC score and immune checkpoint expression, metabolic alterations, and genomic mutations was investigated. RESULTS The TIIC-based model demonstrated superior predictive performance for patient outcomes compared to 53 published models. High TIIC feature score correlated with increased immune infiltration, inflammatory responses, and poor survival. In contrast, low score was associated with enhanced responses to immune checkpoint inhibitors. Significant metabolic reprogramming, including lipid and sulfur metabolism, and distinct genomic alterations, such as BAP1 mutations, were linked to TIIC score. CONCLUSION Our findings underscore the pivotal role of TIIC-RNAs in mediating drug resistance in ccRCC. The prognostic model provides valuable insights into immune and metabolic mechanisms underlying therapy resistance, offering a foundation for developing precision therapeutics targeting the TME.
Collapse
Affiliation(s)
- Jifeng Yang
- School of Clinical Medicine, Hebei University, Affiliated Hospital of Hebei University, Baoding, 071000, China
- Hebei Provincial Key Laboratory of Skeletal Metabolic Physiology of Chronic Kidney Disease, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Yixuan Xing
- Department of Emergency, Xiangya Hospital, Central South University, Changsha, 410031, China
| | - Jiusong Luan
- Pulmonary and Critial Care Medicine, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Wenbo Yang
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xin Zhang
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, 071000, China.
| | - Yanhua Tian
- Second Department of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| | - Haisong Zhang
- School of Clinical Medicine, Hebei University, Affiliated Hospital of Hebei University, Baoding, 071000, China.
- Hebei Provincial Key Laboratory of Skeletal Metabolic Physiology of Chronic Kidney Disease, Affiliated Hospital of Hebei University, Baoding, 071000, China.
| |
Collapse
|
43
|
Chen Y, Huang C, Wei W. Establishment of liquid-liquid phase separation-related prognostic model in lung adenocarcinoma and systematic analysis of its clinical significance. Int J Biol Markers 2025; 40:12-23. [PMID: 39791348 DOI: 10.1177/03936155241310887] [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: 01/12/2025]
Abstract
PurposeTo detect the prognostic importance of liquid-liquid phase separation (LLPS) in lung adenocarcinoma.MethodsThe gene expression files, copy number variation data, and clinical data were downloaded from The Cancer Genome Atlas cohort. LLPS-related genes were acquired from the DrLLPS website. The prognostic model based on LLPS was constructed by the Cox regression and LASSO regression analyses after the identification of LLPS-related differentially expressed genes (DEGs). Gene Ontology functional and Kyoto Encyclopedia of Genes and Genomes enrichment analysis were performed. The LLPS-related prognostic risk score was validated by GSE31210 and GSE72094. The overall survival of lung adenocarcinoma patients was predicted by plotting a nomogram. The biological features of the high-risk lung adenocarcinoma were evaluated by the CIBERSORT, ESTIMATE, Gene Set Variation Analysis, and Genomics of Drug Sensitivity in Cancer. Reverse transcription-quantitative polymerase chain reaction detected hub gene expression.ResultsA total of 91 DEGs were screened out in LLPS, among which 9 genes were discovered as prognostic biomarkers of lung adenocarcinoma. GRIA1, CRTAC1, MAGEA4, and MAPK4 were identified as hub genes by the LASSO Cox regression analysis. High-risk and low-risk groups were divided according to the risk index, with the high-risk group displaying a markedly worse outcome. CRTAC1 expression was significantly decreased, MAGEA4 and MAPK4 expressions were increased, while GRIA1 expression was altered in lung adenocarcinoma cells. Tumor microenvironment, signaling pathway enrichment, and drug sensitivity significantly differed between different risk groups.ConclusionsThis work proposed a prognostic tool based on the LLPS-related gene signature to offer prospective and effective biomarkers for lung adenocarcinoma prognosis.
Collapse
Affiliation(s)
- Yan Chen
- Department of Respiratory and Critical Care Medicine, Anyue County People's Hospital, Anyue, China
| | - Cheng Huang
- Department of Respiratory and Critical Care Medicine, Anyue County People's Hospital, Anyue, China
| | - Wei Wei
- Department of Respiratory and Critical Care Medicine, Anyue County People's Hospital, Anyue, China
| |
Collapse
|
44
|
Cohen R, Raeisi M, Chibaudel B, Yothers G, Goldberg RM, Bachet JB, Wolmark N, Yoshino T, Schmoll HJ, Haller DG, Kerr R, Lonardi S, George TJ, Shacham-Shmueli E, Shi Q, André T, de Gramont A. Impact of tumor and node stages on the efficacy of adjuvant oxaliplatin-based chemotherapy in stage III colon cancer patients: an ACCENT pooled analysis. ESMO Open 2025; 10:104481. [PMID: 40043353 PMCID: PMC11928968 DOI: 10.1016/j.esmoop.2025.104481] [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: 11/27/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Standard adjuvant treatment of stage III colon cancer (CC) is fluoropyrimidine with oxaliplatin. Recently, stage III was subdivided into low-risk (T1-3, N1) and high-risk (T4 and/or N2), with the benefit of adding oxaliplatin varying across these substages. In this study, we aimed to assess the impact of oxaliplatin on survival outcomes in subdividing stage III CC patients based on T and N staging. PATIENTS AND METHODS A total of 4942 stage III CC patients were pooled from the three randomized pivotal trials of oxaliplatin. Kaplan-Meier curves, Cox models stratified by study, and interaction tests were used to assess the oxaliplatin effect across subgroups based on T and N stages. The primary endpoint was overall survival (OS). RESULTS The prevalence of tumor stages was T1-2 12.4%, T3 74.4%, and T4 13.1%; nodal stages were N1 64.7% and N2 35.3%. A significant OS benefit from oxaliplatin was seen only in T3 (5-year OS = 77.2% versus 73.0%, P < 0.001): T3N1 (hazard ratio 0.72, 95% confidence interval 0.62-0.85, P < 0.001) and T3N2 (hazard ratio 0.81, 95% confidence interval 0.69-0.95, P = 0.010). No benefit was observed for T1-2 (5-year OS = 87.8% versus 88.7%, P = 0.644) or T4 patients (5-year OS = 62.6% versus 60.2%, P = 0.648). Subgroup analysis revealed a significant interaction between T stage and the effect of oxaliplatin treatment on OS, whereas no such interaction was observed for N stage. CONCLUSIONS Our analysis revealed that oxaliplatin-based chemotherapy offers a significant survival benefit in stage III CC patients with T3 tumors. In contrast, no survival benefit was observed for T1-2 or T4 patients. These results suggested that T stage plays a more crucial role than N stage in predicting treatment benefit, highlighting the need for tailored treatment strategies based on tumor characteristics.
Collapse
Affiliation(s)
- R Cohen
- Sorbonne University, Department of Medical Oncology, Hôpital Saint-Antoine, AP-HP, and INSERM UMRS 938, Équipe Instabilité des Microsatellites et Cancer, Équipe Labellisée par la Ligue Nationale Contre le Cancer, SIRIC CURAMUS, Centre de recherche Saint Antoine, Paris, France
| | - M Raeisi
- Statistical Unit, ARCAD Foundation, Paris, France.
| | - B Chibaudel
- Department of Medical Oncology, Franco-British Hospital, Levallois-Perret, France
| | - G Yothers
- Department of Biostatistics, University of Pittsburgh and NRG Oncology, Pittsburgh, USA. https://twitter.com/GregYothers
| | - R M Goldberg
- Department of Oncology, West Virginia University Cancer Institute, Morgantown, USA
| | - J-B Bachet
- Hepato-gastroenterology and Digestive Oncology Department, Sorbonne University, Pitié Salpêtrière Hospital, APHP, Paris, France
| | | | - T Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - H-J Schmoll
- Division of Clinical Research in Oncology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - D G Haller
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA
| | - R Kerr
- Department of Oncology, University of Oxford, Oxford, UK
| | - S Lonardi
- Medical Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy. https://twitter.com/sara_lonardi1
| | - T J George
- Division of Hematology and Oncology, University of Florida, Gainesville, USA. https://twitter.com/TGeorgeMD
| | - E Shacham-Shmueli
- Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Q Shi
- Department of Quantitative Health Science, Mayo Clinic, Rochester, USA
| | - T André
- Sorbonne University, Department of Medical Oncology, Hôpital Saint-Antoine, AP-HP, and INSERM UMRS 938, Équipe Instabilité des Microsatellites et Cancer, Équipe Labellisée par la Ligue Nationale Contre le Cancer, SIRIC CURAMUS, Centre de recherche Saint Antoine, Paris, France; ARCAD Foundation, Paris, France
| | - A de Gramont
- Department of Medical Oncology, Franco-British Hospital, Levallois-Perret, France; ARCAD Foundation, Paris, France
| |
Collapse
|
45
|
Li D, Bao L, Liu S, Ji K, Xu X, Yuan J, Xia G. Identification and Validation of Molecular Features of the Anoikis Gene-Related Hub Genes in Nasopharyngeal Carcinoma. Appl Biochem Biotechnol 2025; 197:2066-2092. [PMID: 39666232 DOI: 10.1007/s12010-024-05130-y] [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: 11/20/2024] [Indexed: 12/13/2024]
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant tumor originating from nasopharyngeal mucosa. Anoikis, a form of programmed cell death induced by detachment from the extracellular matrix, normally prevents metastasis. Resistance to anoikis in cancer cells can enhance their metastatic potential. This study identifies anoikis-related genes (ARGs) associated with NPC to elucidate tumorigenesis mechanisms. Analysis of the GSE12452 dataset from GEO revealed 77 differentially expressed ARGs in NPC tissues. GO and KEGG analyses highlighted significant enrichment in apoptosis-related pathways. A PPI network identified MYC, FN1, BRCA1, and FGF2 as Hub genes. Correlation analysis showed MYC positively correlated with activated dendritic cells (p < 0.01) but negatively with naive CD4 T cells (p < 0.001). FN1 was positively correlated with activated dendritic cells (p < 0.01) and negatively with M1 macrophages (p < 0.05). FGF2 negatively correlated with naive CD4 T cells (p < 0.001), while BRCA1 was positively correlated with eosinophils (p < 0.01). GSVA and GSEA indicated that MYC, FN1, BRCA1, and FGF2 were significantly enriched in cell cycle and DNA replication pathways. Immunohistochemistry and qPCR of 50 NPC samples confirmed the overexpression of these genes. Knockdown of MYC, FN1, BRCA1, and FGF2 led to increased tumor cell malignancy, with statistical significance (p < 0.05). This study identifies MYC, FN1, BRCA1, and FGF2 as anoikis-related genes (ARGs) with significant regulatory roles in nasopharyngeal carcinoma (NPC). These ARGs are found to be involved in the development and progression of NPC, suggesting their potential as therapeutic targets for this cancer.
Collapse
Affiliation(s)
- Dong Li
- Department of Otolaryngology, Beilun District People's Hospital of Ningbo, Beilun District, No. 1288, Lushan East Road, Ningbo City, China
| | - Lihao Bao
- Department of Otolaryngology, Beilun District People's Hospital of Ningbo, Beilun District, No. 1288, Lushan East Road, Ningbo City, China
| | - Shaosheng Liu
- Department of Otolaryngology, Beilun District People's Hospital of Ningbo, Beilun District, No. 1288, Lushan East Road, Ningbo City, China
| | - Ke Ji
- Department of Otolaryngology, Beilun District People's Hospital of Ningbo, Beilun District, No. 1288, Lushan East Road, Ningbo City, China
| | - Xujiu Xu
- Department of Otolaryngology, Beilun District People's Hospital of Ningbo, Beilun District, No. 1288, Lushan East Road, Ningbo City, China
| | - Jie Yuan
- Department of Otolaryngology, Beilun District People's Hospital of Ningbo, Beilun District, No. 1288, Lushan East Road, Ningbo City, China
| | - Guihua Xia
- Department of Otolaryngology, Beilun District People's Hospital of Ningbo, Beilun District, No. 1288, Lushan East Road, Ningbo City, China.
| |
Collapse
|
46
|
Jing Y, Wang Y, Li Y, Huang X, Wang J, Yelihamu D, Guo C. Diagnostics and immunological function of CENPN in human tumors: from pan-cancer analysis to validation in breast cancer. Transl Cancer Res 2025; 14:881-906. [PMID: 40104708 PMCID: PMC11912047 DOI: 10.21037/tcr-24-1291] [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: 09/25/2024] [Accepted: 01/03/2025] [Indexed: 03/20/2025]
Abstract
Background Centromere protein N (CENPN), a member of the centromere protein family, contributes to ribonucleic assembly, mitosis progression, and chromosome separation. CENPN manifests a close link with the occurrence and progression of several malignant cancers, but there is no pan-cancer study on CENPN, and we aim to ascertain the connection between CENPN and human cancer prognosis and immunotherapy. Methods The CENPN function in multiple malignant tumors was comprehensively investigated with data from The Cancer Genome Atlas (TCGA) and integrated Gene Expression Omnibus (GEO) database. We examined the transcriptional level, prognostic effect, diagnostic value, genetic and epigenetic alteration, methylation level, and immunological importance of CENPN. Furthermore, this work provided further confirmation of the phenotypic regulating function of CENPN in breast cancer (BC) cells. Results CENPN exhibited significant upregulation in diverse cancer tissues and had different expression patterns across immunological and molecular subgroups in several cancer types. Elevated expression of CENPN may correlate with a worse prognosis. CENPN effectively differentiates most cancers from healthy tissues. Hypomethylate was shown to be CENPN promoter in most cancers. CENPN was shown to be connected with levels of different immune cell infiltration. Kyoto Encyclopedia of Genes and Genomes (KEGG) and the Gene Set Enrichment Analysis (GSEA) analysis suggested that CENPN may mediate neutrophil extranuclear trap formation, cell cycle, and P53 signaling pathways in cancer. In vitro studies showed that the overexpression of CENPN promotes the proliferation, invasion, and migration of BC cells, while concurrently inhibiting their apoptosis. Conclusions CENPN may operate as a novel predictive indicator and molecular target for targeted therapy in pan-cancer. Significantly, CENPN contributed to controlling the BC growth and advancement.
Collapse
Affiliation(s)
- Yubo Jing
- Department of Breast Surgery, Center of Digestive and Vascular, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yiyang Wang
- Department of Breast Surgery, Center of Digestive and Vascular, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yongxiang Li
- Department of Breast Surgery, Center of Digestive and Vascular, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xinzhu Huang
- Department of Breast Surgery, Center of Digestive and Vascular, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Junyi Wang
- Department of Breast Surgery, Center of Digestive and Vascular, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Dlraba Yelihamu
- Department of Breast Surgery, Center of Digestive and Vascular, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Chenming Guo
- Department of Breast Surgery, Center of Digestive and Vascular, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
47
|
Chen X, Yuan Y, Zhou F, Li L, Pu J, Jiang X. m6A RNA methylation: a pivotal regulator of tumor immunity and a promising target for cancer immunotherapy. J Transl Med 2025; 23:245. [PMID: 40022120 PMCID: PMC11871626 DOI: 10.1186/s12967-025-06221-y] [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: 09/27/2024] [Accepted: 02/11/2025] [Indexed: 03/03/2025] Open
Abstract
M6A modification is one of the most common regulatory mechanisms of gene expression in eukaryotic cells, influencing processes such as RNA splicing, degradation, stability, and protein translation. Studies have shown that m6A methylation is closely associated with tumorigenesis and progression, and it plays a key regulatory role in tumor immune responses. m6A modification participates in regulating the differentiation and maturation of immune cells, as well as related anti-tumor immune responses. In the tumor microenvironment, m6A modification can also affect immune cell recruitment, activation, and polarization, thereby promoting or inhibiting tumor cell proliferation and metastasis, and reshaping the tumor immune microenvironment. In recent years, immunotherapies for tumors, such as immune checkpoint inhibitors and adoptive cell immunotherapy, have been increasingly applied in clinical settings, achieving favorable outcomes. Targeting m6A modifications to modulate the immune system, such as using small-molecule inhibitors to target dysregulated m6A regulatory factors or inducing immune cell reprogramming, can enhance anti-tumor immune responses and strengthen immune cell recognition and cytotoxicity against tumor cells. m6A modification represents a new direction in tumor immunotherapy with promising clinical potential. This review discusses the regulatory role of m6A methylation on immune cells and tumor immune responses and explores new strategies for immunotherapy.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Yixiao Yuan
- Department of Medicine, UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | - Fan Zhou
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Lihua Li
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Jun Pu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China.
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China.
| | - Xiulin Jiang
- Department of Medicine, UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
48
|
Zhang YT, Wei YN, Liu CC, Yang MQ. Bibliometric analysis: a study of the microenvironment in cervical cancer (2000-2024). Front Oncol 2025; 15:1508173. [PMID: 40083880 PMCID: PMC11903265 DOI: 10.3389/fonc.2025.1508173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025] Open
Abstract
Objective The incidence of cervical cancer has increased in recent years. The tumor microenvironment (TME) is the local biological environment involved in tumor occurrence and development. This study aimed to conduct a comprehensive analysis of the global research on the TME in cervical cancer (CC), providing a knowledge framework in this field from a holistic and systematic perspective based on a bibliometric analysis. Methods Studies focusing on the TME in cervical cancer were searched using the Web of Science Core Collection database. The annual output, cooperation, hotspots, research status, and development trends in this field were analyzed using bibliometric softwares (VOSviewer and CiteSpace). Results A total of 1,057 articles published between 2000 and 2024 were selected. The number of publications and citations has recently increased. Cooperation network analysis indicated that China holds the foremost position in research on the TME in cervical cancer with the highest volume of publications, thus exerting the greatest influence. Fudan University had the highest output. Frontiers in Oncology showed the highest degree of productivity in this field. Rofstad, Einar K. made the most article contributions and was the most co-cited author. Four clusters were obtained after a cluster analysis of the keywords: TME, cervical cancer, immunotherapy, and prognosis. Immunotherapy, human papillomavirus, and biomarkers were relatively recent keywords that attracted increasing attention from researchers. Discussion This bibliometric analysis provides a data-based and objective introduction to the TME of cervical cancer, and offers readers a valuable reference for future research. Conclusions Comprehensive research in this field was mainly distributed in the TME of cervical cancer through the analysis of keywords and documents. Sufficient evidence supports mechanism research and application exploration. Further research should explore new topics related to the TME of cervical cancer.
Collapse
Affiliation(s)
- Yun-Tao Zhang
- Department of Obstetrics, Changyi People’s Hospital, Changyi, Shandong, China
| | - Yan-Ni Wei
- Faculty of Health Management, Weifang Nursing Vocational College, Weifang, Shandong, China
| | - Chen-Chen Liu
- Department of Pathology, Weifang People’s Hospital (First Affiliated Hospital of Shandong Second Medical University), Weifang, Shandong, China
| | - Mai-Qing Yang
- Department of Pathology, Weifang People’s Hospital (First Affiliated Hospital of Shandong Second Medical University), Weifang, Shandong, China
| |
Collapse
|
49
|
Vallieri N, Datsi A. Immune Cell Interplay in the Fight Against GBM. Cancers (Basel) 2025; 17:817. [PMID: 40075663 PMCID: PMC11899300 DOI: 10.3390/cancers17050817] [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: 01/26/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Despite multimodal therapies, the treatment of glioblastoma remains challenging. In addition to the very complex mechanisms of cancer cells, including specialized phenotypes that enable them to proliferate, invade tissues, and evade immunosurveillance, they exhibit a pronounced resistance to chemo- and radiotherapy. More advanced tumors create a hypoxic environment that supports their proliferation and survival, while robust angiogenesis ensures a constant supply of nutrients. In GBM, these structures are very pronounced and contribute to the creation and maintenance of a highly immunosuppressive microenvironment that promotes tumor growth and immune escape. In addition, the high accumulation of immunosuppressive tumor-infiltrating leukocytes and other cells, the pronounced expression of immune checkpoint molecules, and the low mutational burden, i.e., the low number of neoantigens, are hallmarks of GBM and contribute to the challenge of therapeutic approaches. Here, we review a number of mechanisms that GBM exploits to support tumor growth and potential treatments. These include new chemotherapeutics, tumor treating fields, and small molecules, including compounds targeting angiogenesis or blockers of tyrosine kinases that inhibit tumor cell proliferation and survival. In addition, we focus on immunotherapies such as immune checkpoint blockade or cell therapies, in particular vaccination with dendritic cells and CAR-T cells, which can either kill GBM cells directly or bypass immunosuppression by modulating the tumor microenvironment or boosting the patient's own immune response.
Collapse
Affiliation(s)
| | - Angeliki Datsi
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, 40225 Duesseldorf, Germany;
| |
Collapse
|
50
|
Kuang J, Li J, Zhou S, Li Y, Lin J, Huang W, Yuan X. Genomic and micro-environmental insights into drug resistance in colorectal cancer liver metastases. Discov Oncol 2025; 16:241. [PMID: 40009285 DOI: 10.1007/s12672-025-01976-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is known for its high heterogeneity, with liver metastases significantly impairing survival outcomes. Understanding the tumor microenvironment (TME) and genomic alterations in metastatic sites is crucial for developing personalized therapies that overcome drug resistance and improve prognosis. METHODS We profiled 54 CRC liver metastases, comparing them with 198 other metastatic lesions and normal liver tissues. By analyzing immune cell infiltration, stromal interactions, and key genomic alterations, we constructed an 11-gene prognostic model to predict survival and immunotherapy outcomes. RESULTS CRC liver metastases with high-risk profiles demonstrated enriched follicular helper T cells, activated dendritic cells, and M2 macrophages in the TME. Frequent mutations in APC, TP53, KRAS, and PIK3CA were identified, alongside altered EGFR signaling. The 11-gene model effectively stratified patients by prognosis and predicted immunotherapy responses, emphasizing the therapeutic potential of targeting resistance mechanisms. CONCLUSIONS This study reveals how genomic and TME-driven factors contribute to drug resistance in CRC liver metastases. Integrating these insights with clinical data could advance precision therapies, addressing the evolving challenge of tumor drug resistance in CRC.
Collapse
Affiliation(s)
- Junjie Kuang
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Jun Li
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Siwei Zhou
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Yi Li
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Jinbo Lin
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Weizhen Huang
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China.
| | - Xia Yuan
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China.
| |
Collapse
|