1
|
Xie Y, Mi X, Xing Y, Dai Z, Pu Q. Past, present, and future of exosomes research in cancer: A bibliometric and visualization analysis. Hum Vaccin Immunother 2025; 21:2488551. [PMID: 40207548 PMCID: PMC11988232 DOI: 10.1080/21645515.2025.2488551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer seriously threatens the lives and health of people worldwide, and exosomes seem to play an important role in managing cancer effectively, which has attracted extensive attention from researchers in recent years. This study aimed to scientifically visualize exosomes research in cancer (ERC) through bibliometric analysis, reviewing the past, summarizing the present, and predicting the future, with a view to providing valuable insights for scholars and policy makers. Researches search and data collection from Web of Science Core Collection and clinical trial.gov. Calculations and visualizations were performed using Microsoft Excel, VOSviewer, Bibliometrix R-package, and CiteSpace. As of December 1, 2024, and March 8, 2025, we identified 8,001 ERC-related publications and 107 ERC-related clinical trials, with an increasing trend in annual publications. Our findings supported that China, Nanjing Medical University, and International Journal of Molecular Sciences were the most productive countries, institutions, and journals, respectively. Whiteside, Theresa L. had the most publications, while Théry, C was the most co-cited scholar. In addition, Cancer Research was the most co-cited journal. Spatial and temporal distribution of clinical trials was the same as for publications. High-frequency keywords were "extracellular vesicle," "microRNA" and "biomarker." Additional, "surface functionalization," "plant," "machine learning," "nanomaterials," "promotes metastasis," "engineered exosomes," and "macrophage-derived exosomes" were promising research topics. Our study comprehensively and visually summarized the structure, hotspots, and evolutionary trends of ERC. It would inspire subsequent studies from a macroscopic perspective and provide a basis for rational allocation of resources and identification of collaborations among researchers.
Collapse
Affiliation(s)
- Yafei Xie
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Xingqi Mi
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yikai Xing
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Zhangyi Dai
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Qiang Pu
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Gupta M, Verma N, Sharma N, Singh SN, Brojen Singh RK, Sharma SK. Deep transfer learning hybrid techniques for precision in breast cancer tumor histopathology classification. Health Inf Sci Syst 2025; 13:20. [PMID: 39949707 PMCID: PMC11813847 DOI: 10.1007/s13755-025-00337-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 01/07/2025] [Indexed: 02/16/2025] Open
Abstract
The breast cancer is one of the most prevalent causes of cancer-related death globally. Preliminary diagnosis of breast cancer increases the patient's chances of survival. Breast cancer classification is a challenging problem due to dense tissue structures, subtle variations, cellular heterogeneity, artifacts, and variability. In this paper, we propose three hybrid deep-transfer learning models for breast cancer classification using histopathology images. These models use Xception model as a base model, and we add seven more layers to fine-tune the base model. We also performed an extensive comparative analysis of five prominent machine-learning classifiers, namely Random Forest Classifier (RFC), Logistic Regression (LR), Support Vector Classifier (SVC), K-Nearest Neighbors (KNN), and Ada-boost. We incorporate the best performing two classifiers, namely RFC and SVC, in the fine-tuned Xception model, and accordingly, they are named as Xception Random Forest (XRF) and Xception Support Vector (XSV), respectively. The fine-tuned Xception model with softmax classifier is termed as Multi-layer Xception Classifier (MXC). These three models are evaluated on the two publically available datasets: BreakHis and Breast Histopathology Images Database (BHID). Our all three models perform better than the state-of-the-art methods. The XRF provides the best performance at the 40 × magnification level on the BreakHis dataset, with an accuracy (ACC) of 94.44%, F1 score (F1) of 94.44%, area under the receiver operating characteristic curve (AUC) of 95.12%, Matthew's correlation coefficient (MCC) of 88.98%, kappa (K) of 88.88%, and classification success index (CSI) of 89.23%. The MXC provides the best performance on the BHID dataset, with an ACC of 88.50%, F1 of 88.50%, AUC of 95.12%, MCC of 77.03%, K of 77.00%, and CSI of 79.13%. Further, to validate our models, we performed fivefold cross-validation on both datasets and obtained similar results.
Collapse
Affiliation(s)
- Muniraj Gupta
- School of Computer & Systems Sciences, Jawaharlal Nehru University, New Delhi, 110067 India
| | - Nidhi Verma
- Ramlal Anand College, University of Delhi, South Campus, Anand Niketan, New Delhi, 110021 India
| | - Naveen Sharma
- Indian Council of Medical Research, New Delhi, 110029 India
| | | | - R. K. Brojen Singh
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi, 110067 India
| | - Saurabh Kumar Sharma
- School of Computer & Systems Sciences, Jawaharlal Nehru University, New Delhi, 110067 India
| |
Collapse
|
3
|
Wang Q, Sun N, Zhang C, Kunzke T, Zens P, Feuchtinger A, Berezowska S, Walch A. Metabolic heterogeneity in tumor cells impacts immunology in lung squamous cell carcinoma. Oncoimmunology 2025; 14:2457797. [PMID: 39924768 PMCID: PMC11812363 DOI: 10.1080/2162402x.2025.2457797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/22/2024] [Accepted: 01/20/2025] [Indexed: 02/11/2025] Open
Abstract
Metabolic processes are crucial in immune regulation, yet the impact of metabolic heterogeneity on immunological functions remains unclear. Integrating metabolomics into immunology allows the exploration of the interactions of multilayered features in the biological system and the molecular regulatory mechanism of these features. To elucidate such insight in lung squamous cell carcinoma (LUSC), we analyzed 106 LUSC tumor tissues. We performed high-resolution matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI) to obtain spatial metabolic profiles, and immunohistochemistry to detect tumor-infiltrating T lymphocytes (TILs). Unsupervised k-means clustering and Simpson's diversity index were employed to assess metabolic heterogeneity, identifying five distinct metabolic tumor subpopulations. Our findings revealed that TILs are specifically associated with metabolite distributions, not randomly distributed. Integrating a validation cohort, we found that heterogeneity-correlated metabolites interact with CD8+ TIL-associated genes, affecting survival. High metabolic heterogeneity was linked to worse survival and lower TIL levels. Pathway enrichment analyses highlighted distinct metabolic pathways in each subpopulation and their potential responses to chemotherapy. This study uncovers the significant impact of metabolic heterogeneity on immune functions in LUSC, providing a foundation for tailoring therapeutic strategies.
Collapse
Affiliation(s)
- Qian Wang
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Na Sun
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Chaoyang Zhang
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Kunzke
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Philipp Zens
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Sabina Berezowska
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Axel Walch
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
4
|
Wei T, Fu G, Zhao J, Cao F, Guo D. Acyl-CoA dehydrogenase long chain acts as a tumor-suppressive factor in lung adenocarcinoma progression. Cell Adh Migr 2025; 19:2495676. [PMID: 40262559 PMCID: PMC12026206 DOI: 10.1080/19336918.2025.2495676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/12/2024] [Accepted: 04/05/2025] [Indexed: 04/24/2025] Open
Abstract
This study investigated the role of long-chain acyl-CoA dehydrogenase (ACADL) in lung adenocarcinoma (LUAD). ACADL was significantly downregulated in human LUAD tissues compared to normal lung tissues. In vitro, ectopic expression of ACADL in murine LLC cells decreased cell viability, migration, and invasion, while ACADL knockdown exhibited the opposite effect. In vivo, ACADL overexpression impeded tumor growth and metastasis. Mechanistically, ACADL hindered tumor progression by inducing cell cycle arrest, promoting apoptosis, and suppressing the epithelial-mesenchymal transition (EMT) process. These findings suggest ACADL acts as a tumor suppressor in LUAD progression.
Collapse
Affiliation(s)
- Tingju Wei
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guowei Fu
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junjie Zhao
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fengan Cao
- Department of Respiratory Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Guo
- Henan Key Laboratory for Digestive Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
5
|
Talukder AK, Naib AA, Mamo S, Thompson L, Ferst JG, Rabaglino MB, Browne JA, Fair T, Lonergan P. Specificity protein 1 (SP1) plays an essential role in early bovine embryo development. Theriogenology 2025; 242:117455. [PMID: 40286422 DOI: 10.1016/j.theriogenology.2025.117455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/22/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
A previous RNA-Seq study revealed that the transcript abundance of specificity protein 1 (SP1) was significantly higher in Day 7 bovine blastocysts compared to conceptuses on Days 10, 13, 16, and 19, suggesting a stage-specific role in early bovine embryo development. The present study aimed to characterize the mRNA expression of SP1 and associated candidate genes (ACSS1, C1QBP, ATF3, MAT2A, and POLD1) during early bovine embryo development from the 2-cell to blastocyst stage. Further, the effects of SP1 inhibition on embryo development were evaluated by culturing embryos with the SP1 inhibitor, mithramycin A (MT) at varying concentrations (0, 25, 50, 100, and 1000 nM). As further validation, we examined expression of SP1 and associated genes by interrogating transcriptomic data from Day 4 (16-cell stage) embryos cultured in vitro or in vivo in the oviducts of lactating or nonlactating dairy cows. The relative abundance of SP1 peaked at the time of embryonic genome activation, being higher (P < 0.05) in 8- and 16-cell embryos compared to the 2-cell stage, and decreasing thereafter (at the morula and blastocyst stages). Similarly, transcript abundance for most of the selected candidate genes involved in the SP1 network were upregulated (P < 0.05) at the 8- and 16-cell stage, but not at other stages investigated. Inhibition of SP1 with MT did not affect embryo development up to the 8-cell stage but reduced (P < 0.05) the proportion of embryos reaching the 16-cell and blastocyst stages in a dose-dependent manner. Moreover, blastocysts produced in the presence of MT contained fewer (P < 0.05) cells than blastocysts developed without MT. Expression of SP1 and associated genes in 16-cell stage (Day 4) embryos produced either in vitro or in vivo was higher (P < 0.05) compared to in vitro-produced 2- to 4- cell stage (Day 2) embryos. These findings suggest an essential role of SP1 during early embryo development, particularly around the time of embryonic genomic activation.
Collapse
Affiliation(s)
- Anup K Talukder
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland; Department of Gynecology, Obstetrics and Reproductive Health, Gazipur Agricultural University, Gazipur, 1706, Bangladesh
| | - Abdullah A Naib
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Solomon Mamo
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Laura Thompson
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Juliana G Ferst
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Maria B Rabaglino
- Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Netherlands
| | - John A Browne
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Trudee Fair
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Pat Lonergan
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
6
|
Xi J, Liu Y, Zhang L, Zhang B, Zhao J, Fang J. Redox dyshomeostasis-driven prodrug strategy for enhancing camptothecin-based chemotherapy: Selenization of SN38 as a case study. Bioorg Chem 2025; 160:108468. [PMID: 40245475 DOI: 10.1016/j.bioorg.2025.108468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/01/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025]
Abstract
Harnessing the modulation of redox homeostasis represents a promising anticancer strategy. Here, we design and evaluate Se-SN38, a prodrug of the camptothecin (CPT) derivative 7-ethyl-10-hydroxycamptothecin (SN38) with a cyclic five-membered diselenide moiety for redox-triggered activation. We demonstrate that Se-SN38 exhibits superior cytotoxicity in various cancer cell lines over the parent drug SN38 or the control prodrug S-SN38, a sulfur analogue of Se-SN38. This increased potency is attributed to the efficient release of SN38 and induction of oxidative stress, as demonstrated by a significant rise in reactive oxygen species production, along with a marked depletion of cellular total thiols and a decreased GSH/GSSG ratio. Furthermore, Se-SN38 treatment leads to inhibition of thioredoxin reductase activity, disruption of mitochondrial membrane potential, and induction of DNA damage, culminating in apoptosis. These findings suggest that Se-SN38 represents a promising strategy to enhance the therapeutic efficacy of CPT derivatives by exploiting the unique redox-active properties of cyclic five-membered diselenide to induce oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Junmin Xi
- School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, Jiangsu 210094, China
| | - Yu Liu
- School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, Jiangsu 210094, China
| | - Linjie Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Jintao Zhao
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China.
| | - Jianguo Fang
- School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, Jiangsu 210094, China.
| |
Collapse
|
7
|
Goyal A, Afzal M, Khan NH, Goyal K, Srinivasamurthy SK, Gupta G, Benod Kumar K, Ali H, Rana M, Wong LS, Kumarasamy V, Subramaniyan V. Targeting p53-p21 signaling to enhance mesenchymal stem cell regenerative potential. Regen Ther 2025; 29:352-363. [PMID: 40248767 PMCID: PMC12004386 DOI: 10.1016/j.reth.2025.03.007] [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: 01/26/2025] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/19/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are properties of self-renewal and differentiation potentials and thus are very appealing to regenerative medicine. Nevertheless, their therapeutic potential is frequently constrained by senescence, limited proliferation, and stress-induced apoptosis. The key role of the p53-p21 biology in MSC biology resides in safeguarding genomic stability while promoting senescence and limiting regenerative capacity upon over-activation demonstrated. This pathway is a key point for improving MSC function and exploiting the inherent limitations. Recent advances indicate that senescence can be delayed by targeting the p53-p21 signaling and improved MSC proliferation and differentiation capacity. PFT-α pharmacological agents transiently inhibit p53 from increasing proliferation and lineage-specific differentiation, while antioxidants such as hydrogen-rich saline and epigallocatechin 3 gallate (EGCG) suppress oxidative stress and attenuate p53 p21 signaling. Genetic tools like CRISPR-Cas9 and RNA interference also precisely modulate TP53 and CDKN1A expression to optimize MSC functionality. The interplay of p53-p21 with pathways like Wnt/β-catenin and MAPK further highlights opportunities for combinatorial therapies to enhance MSC resilience and regenerative outcomes. This review aims to offer a holistic view of how p53-p21 targeting can further the regenerative potential of MSCs, resolving senescence, proliferation, and stress resilience towards advanced therapeutics built on MSCs.
Collapse
Affiliation(s)
- Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Suresh Kumar Srinivasamurthy
- Department of Pharmacology, Ras Al Khaimah College of Medical Sciences, Ras Al Khaimah Medical & Health Sciences University, P.O. Box 11172, Ras Al Khaimah, United Arab Emirates
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - K. Benod Kumar
- Department of General Surgery, Consultant Head and Neck Surgical Oncology, Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai 71800, Malaysia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Division of Pharmacology, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia
| |
Collapse
|
8
|
Chen W, Zhang Z, Han Y, Li X, Liu C, Sun Y, Ren Y, Guan X. Remodeling tumor microenvironment by versatile nanoplatform orchestrated mechanotherapy with chemoimmunotherapy to synergistically enhance anticancer efficiency. Biomaterials 2025; 317:123104. [PMID: 39813969 DOI: 10.1016/j.biomaterials.2025.123104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
Solid tumors (particularly the desmoplastic ones) usually harbor insurmountable mechanical barriers and formidable immunosuppressive tumor microenvironment (TME), which severely restricted nanomedicine-penetration and vastly crippled outcomes of numerous therapies. To overcome these barriers, a versatile nanoplatform orchestrated mechanotherapy with chemoimmunotherapy was developed here to simultaneously modulate tumor physical barriers and remodel TME for synergistically enhancing anticancer efficiency. Dexamethasone (DMS) and cis-aconityl-doxorubicin (CAD) were co-hitchhiked into phenylboronic acid functionalized polyethylenimine (PEI-PBA) carrier, and further in situ shielded by aldehyde-modified polyethylene glycol (PEG) to form CAD/DMS@PEG/PEI-PBA (CD@PB) nanoparticles (NPs). The CD@PB NPs exhibited multifunctionality: (1) Long in vivo circulation and acidic TME-responsive PEG deshielding for being efficiently internalized into cells. (2) Endosomal-pH triggered drug release and PEI-facilitated drug-escaping from endosome into cytoplasm. (3) DMS down-regulated thick stroma and weakened mechanical barriers for facilitating NP penetration. (4) DMS mediated nuclear pore dilation to promote more DOX entering nucleus and enhance treatment effects. (5) DOX induced potent immunogenic cell death (ICD), activated antitumor immunity and exerted chemoimmunotherapy. The versatile CD@PB NPs displayed excellent antitumor efficacy against 4T1 mouse breast cancer, which effectively remodeled immunosuppressive TME and orchestrated mechanotherapy with chemoimmunotherapy to synergistically enhance antitumor efficiency. The study provided an illuminating paradigm for multidimensional cancer therapy.
Collapse
Affiliation(s)
- Wenqiang Chen
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China
| | - Zhe Zhang
- School of Biological and Chemical Engineering, Panzhihua University, Panzhihua, 617000, China
| | - Yunfei Han
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China
| | - Xinyu Li
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China
| | - Chunhui Liu
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China
| | - Yanju Sun
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China
| | - Yanyan Ren
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China
| | - Xiuwen Guan
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, China.
| |
Collapse
|
9
|
Zhang M, Liu C, Tu J, Tang M, Ashrafizadeh M, Nabavi N, Sethi G, Zhao P, Liu S. Advances in cancer immunotherapy: historical perspectives, current developments, and future directions. Mol Cancer 2025; 24:136. [PMID: 40336045 DOI: 10.1186/s12943-025-02305-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/15/2025] [Indexed: 05/09/2025] Open
Abstract
Cancer immunotherapy, encompassing both experimental and standard-of-care therapies, has emerged as a promising approach to harnessing the immune system for tumor suppression. Experimental strategies, including novel immunotherapies and preclinical models, are actively being explored, while established treatments, such as immune checkpoint inhibitors (ICIs), are widely implemented in clinical settings. This comprehensive review examines the historical evolution, underlying mechanisms, and diverse strategies of cancer immunotherapy, highlighting both its clinical applications and ongoing preclinical advancements. The review delves into the essential components of anticancer immunity, including dendritic cell activation, T cell priming, and immune surveillance, while addressing the challenges posed by immune evasion mechanisms. Key immunotherapeutic strategies, such as cancer vaccines, oncolytic viruses, adoptive cell transfer, and ICIs, are discussed in detail. Additionally, the role of nanotechnology, cytokines, chemokines, and adjuvants in enhancing the precision and efficacy of immunotherapies were explored. Combination therapies, particularly those integrating immunotherapy with radiotherapy or chemotherapy, exhibit synergistic potential but necessitate careful management to reduce side effects. Emerging factors influencing immunotherapy outcomes, including tumor heterogeneity, gut microbiota composition, and genomic and epigenetic modifications, are also examined. Furthermore, the molecular mechanisms underlying immune evasion and therapeutic resistance are analyzed, with a focus on the contributions of noncoding RNAs and epigenetic alterations, along with innovative intervention strategies. This review emphasizes recent preclinical and clinical advancements, with particular attention to biomarker-driven approaches aimed at optimizing patient prognosis. Challenges such as immunotherapy-related toxicity, limited efficacy in solid tumors, and production constraints are highlighted as critical areas for future research. Advancements in personalized therapies and novel delivery systems are proposed as avenues to enhance treatment effectiveness and accessibility. By incorporating insights from multiple disciplines, this review aims to deepen the understanding and application of cancer immunotherapy, ultimately fostering more effective and widely accessible therapeutic solutions.
Collapse
Affiliation(s)
- Meiyin Zhang
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chaojun Liu
- Department of Breast Surgery, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University; People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Jing Tu
- Department of Pulmonary and Critical Care Medicine, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Min Tang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, V8 V 1P7, Canada
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR) Yong Loo Lin, School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Peiqing Zhao
- Translational Medicine Center, Zibo Central Hospital Affiliated to Binzhou Medical University, No. 54 Communist Youth League Road, Zibo, China.
| | - Shijian Liu
- Department of General Medicine, The 2nd Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Harbin, 150081, China.
| |
Collapse
|
10
|
Gori K, Baez-Ortega A, Strakova A, Stammnitz MR, Wang J, Chan J, Hughes K, Belkhir S, Hammel M, Moralli D, Bancroft J, Drydale E, Allum KM, Brignone MV, Corrigan AM, de Castro KF, Donelan EM, Faramade IA, Hayes A, Ignatenko N, Karmacharya R, Koenig D, Lanza-Perea M, Lopez Quintana AM, Meyer M, Neunzig W, Pedraza-Ordoñez F, Phuentshok Y, Phuntsho K, Ramirez-Ante JC, Reece JF, Schmeling SK, Singh S, Tapia Martinez LJ, Taulescu M, Thapa S, Thapa S, van der Wel MG, Wehrle-Martinez AS, Stratton MR, Murchison EP. Horizontal transfer of nuclear DNA in transmissible cancer. Proc Natl Acad Sci U S A 2025; 122:e2424634122. [PMID: 40261943 DOI: 10.1073/pnas.2424634122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/05/2025] [Indexed: 04/24/2025] Open
Abstract
Horizontal transfer of nuclear DNA between cells of host and cancer is a potential source of adaptive variation in cancer cells. An understanding of the frequency and significance of this process in naturally occurring tumors is, however, lacking. We screened for this phenomenon in the transmissible cancers of dogs and Tasmanian devils and found an instance in the canine transmissible venereal tumor (CTVT). This involved introduction of a 15-megabase dicentric genetic element, composed of 11 fragments of six chromosomes, to a CTVT sublineage occurring in Asia around 2,000 y ago. The element forms the short arm of a small submetacentric chromosome and derives from a dog with ancestry associated with the ancient Middle East. The introduced DNA fragment is transcriptionally active and has adopted the expression profile of CTVT. Its features suggest that it may derive from an engulfed apoptotic body. Our findings indicate that nuclear horizontal gene transfer, although likely a rare event in tumor evolution, provides a viable mechanism for the acquisition of genetic material in naturally occurring cancer genomes.
Collapse
Affiliation(s)
- Kevin Gori
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Adrian Baez-Ortega
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton CB10 1SA, United Kingdom
| | - Andrea Strakova
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Maximilian R Stammnitz
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Jinhong Wang
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Jonathan Chan
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Katherine Hughes
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Sophia Belkhir
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Maurine Hammel
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Daniela Moralli
- Pandemic Sciences Institute, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - James Bancroft
- Cellular Imaging Core Facility, Centre for Human Genetics, University of Oxford, Oxford OX3 7BM, United Kingdom
| | - Edward Drydale
- Cellular Imaging Core Facility, Centre for Human Genetics, University of Oxford, Oxford OX3 7BM, United Kingdom
| | | | - María Verónica Brignone
- Faculty of Veterinary Sciences, Universidad de Buenos Aires, Buenos Aires C1053ABJ, Argentina
| | - Anne M Corrigan
- School of Veterinary Medicine, St. George's University, True Blue, Grenada
| | - Karina F de Castro
- Faculty of Agrarian and Veterinary Sciences, São Paulo State University, Jaboticabal 14884-900, Brazil
| | - Edward M Donelan
- Animal Management in Rural and Remote Indigenous Communities, Darwin, NT 0820, Australia
| | | | - Alison Hayes
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | | | - Rockson Karmacharya
- Veterinary Diagnostic and Research Laboratory Pvt. Ltd., Kathmandu 44600, Nepal
| | | | - Marta Lanza-Perea
- School of Veterinary Medicine, St. George's University, True Blue, Grenada
| | | | | | | | | | | | | | - Juan C Ramirez-Ante
- Facultad de Ciencias Pecuarias, Corporación Universitaria Santa Rosa de Cabal, Santa Rosa de Cabal 661020, Colombia
| | - John F Reece
- Help in Suffering, Jaipur 302018, Rajasthan, India
| | | | - Sanjay Singh
- Help in Suffering, Jaipur 302018, Rajasthan, India
| | | | - Marian Taulescu
- Department of Anatomic Pathology, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca 400372, Romania
| | - Samir Thapa
- Kathmandu Animal Treatment Centre, Kathmandu 44622, Nepal
| | - Sunil Thapa
- Animal Nepal, Dobighat, Kathmandu 44600, Nepal
| | | | | | - Michael R Stratton
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton CB10 1SA, United Kingdom
| | - Elizabeth P Murchison
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| |
Collapse
|
11
|
Henretta S, Lammerding J. Nuclear envelope proteins, mechanotransduction, and their contribution to breast cancer progression. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:14. [PMID: 40337116 PMCID: PMC12052594 DOI: 10.1038/s44341-025-00018-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 03/18/2025] [Indexed: 05/09/2025]
Abstract
Breast cancer cells frequently exhibit changes in the expression of nuclear envelope (NE) proteins such as lamins and emerin that determine the physical properties of the nucleus and contribute to cellular mechanotransduction. This review explores the emerging interplay between NE proteins, the physical challenges incurred during metastatic progression, and mechanotransduction. Improved insights into the underlying mechanisms may ultimately lead to better prognostic tools and treatment strategies for metastatic breast cancer.
Collapse
Affiliation(s)
- Sarah Henretta
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY USA
| | - Jan Lammerding
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY USA
| |
Collapse
|
12
|
Wang W, Chaudhary R, Szpendyk J, El Khalki L, Yousafzai NA, Chan R, Desai A, Sossey-Alaoui K. Kindlin-2-Mediated Hematopoiesis Remodeling Regulates Triple-Negative Breast Cancer Immune Evasion. Mol Cancer Res 2025; 23:450-462. [PMID: 39918417 DOI: 10.1158/1541-7786.mcr-24-0698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/09/2024] [Accepted: 02/05/2025] [Indexed: 05/03/2025]
Abstract
Triple-negative breast cancer (TNBC) presents significant clinical challenges because of its limited treatment options and aggressive behavior, often associated with poor prognosis. This study focuses on kindlin-2, an adapter protein, and its role in TNBC progression, particularly in hematopoiesis-mediated immune evasion. TNBC tumors expressing high levels of kindlin-2 induce a notable reshaping of hematopoiesis, promoting the expansion of myeloid cells in the bone marrow and spleen. This shift correlated with increased levels of neutrophils and monocytes in tumor-bearing mice over time. Conversely, genetic knockout (KO) of kindlin-2 mitigated this myeloid bias and fostered T-cell infiltration within the tumor microenvironment, indicating the pivotal role of kindlin-2 in immune modulation. Further investigations revealed that kindlin-2 deficiency led to reduced expression of PD-L1, a critical immune checkpoint inhibitor, in TNBC tumors. This molecular change sensitized kindlin-2-deficient tumors to host antitumor immune responses, resulting in enhanced tumor suppression in immunocompetent mouse models. Single-cell RNA sequencing, bulk RNA sequencing, and IHC data supported these findings by highlighting enriched immune-related pathways and increased infiltration of immune cells in kindlin-2-deficient tumors. Therapeutically, targeting PD-L1 in kindlin-2-expressing TNBC tumors effectively inhibited tumor growth, akin to the effects observed with genetic kindlin-2 KO or PD-L1 KO. Our data underscore kindlin-2 as a promising therapeutic target in combination with immune checkpoint blockade to bolster antitumor immunity and counteract resistance mechanisms typical of TNBC and other immune-evasive solid tumors. Implications: Kindlin-2 regulates tumor immune evasion through the systemic modulation of hematopoiesis and PD-L1 expression, which warrants therapeutic targeting of kindlin-2 in patients with TNBC.
Collapse
Affiliation(s)
- Wei Wang
- MetroHealth System, Cleveland, Ohio
- Case Western Reserve University, Cleveland, Ohio
| | | | | | - Lamyae El Khalki
- Case Western Reserve University, Cleveland, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Neelum Aziz Yousafzai
- Case Western Reserve University, Cleveland, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Ricky Chan
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Amar Desai
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Khalid Sossey-Alaoui
- MetroHealth System, Cleveland, Ohio
- Case Western Reserve University, Cleveland, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
| |
Collapse
|
13
|
Liao Q, Jin Z, Long H, Zhu B. Deciphering cancer complexity: perspective on hematopoietic remodeling-mediated immunosuppression. Oncogene 2025; 44:1230-1233. [PMID: 40195467 DOI: 10.1038/s41388-025-03361-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 04/09/2025]
Affiliation(s)
- Qian Liao
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing, China
| | - Zheng Jin
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing, China
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
- Chongqing Key Laboratory of Immunotherapy, Chongqing, China.
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
- Chongqing Key Laboratory of Immunotherapy, Chongqing, China.
| |
Collapse
|
14
|
Lu XY, Zhu LY, Zhu H, Huang SJ, Yang YS, Jiang CR, Ye RR. Cyclometalated iridium(III)-lonidamine conjugates: Mitochondrial targeting and pyroptosis induction. J Inorg Biochem 2025; 266:112852. [PMID: 39938148 DOI: 10.1016/j.jinorgbio.2025.112852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/27/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
A series of cyclometalated Ir(III)-lonidamine (LND) complexes (Ir-LND-1-6) with the formula [Ir(C^N)2bpy(4-CH3-4'-CH2OLND)](PF6) (Ir-LND-1-3) and [Ir(C^N)2bpy(4-CH2OLND-4'-CH2OLND)](PF6) (Ir-LND-4-6) (C^N = 2-phenylpyridine (ppy, in Ir-LND-1 and Ir-LND-4), 2-(2-thienyl) pyridine (thpy, in Ir-LND-2 and Ir-LND-5) and 2-(2,4-difluorophenyl) pyridine (dfppy, in Ir-LND-3 and Ir-LND-6)), were designed and synthesized. 3-(4,5-dimethylthiazol-2-yl)-2,5-biphenyltetrazolium bromide (MTT) assay data showed that the cytotoxicity of Ir-LND-1-3 carry one LND moiety was superior to that of Ir-LND-4-6 with two LND moieties. Therefore, we selected Ir-LND-1-3 as model compounds to investigate the anti-tumor mechanism of the Ir(III)-LND system. The results showed that Ir-LND-1-3 could inhibit cancer cell migration and colony formation. In addition, Ir-LND-1-3 could penetrate into HeLa cells and localized to mitochondria, further disrupting mitochondrial membrane potential (MMP), increasing intracellular reactive oxygen species (ROS), and reducing intracellular adenosine triphosphate (ATP). Further exploration of anti-tumor mechanisms showed that pyroptosis was the main mode of Ir-LND-1-3 induced cell death, manifested as membrane perforation and swelling, activation of caspase-3 and cleavage of Gasdermin E (GSDME), as well as release of lactic dehydrogenase (LDH) and ATP. The pyroptosis induced by Ir-LND-1-3 also initiated immunogenic cell death (ICD) by triggering the release of calreticulin (CRT) and high mobility group protein b1 (HMGB1) on the cell surface.
Collapse
Affiliation(s)
- Xing-Yun Lu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Lin-Yuan Zhu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Hou Zhu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Shao-Jun Huang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China.
| | - Yong-Sheng Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Chun-Rong Jiang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Rui-Rong Ye
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China.
| |
Collapse
|
15
|
Feng ZW, Li L, Zhang SD, Wang YJ, Pei JY, Chen NN, Wu BD, Zheng QL, You QD, Guo XK, Xu XL. Structural Optimization of Pyrazole Compounds as Hsp90 Regulators with Enhanced Antitumor Activity. J Med Chem 2025. [PMID: 40310691 DOI: 10.1021/acs.jmedchem.4c02182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Targeting Hsp90 is an effective strategy for cancer therapy. TAS-116 has been approved for the treatment of gastrointestinal stromal tumors. Our previous studies identified a series of pyrazole derivatives as covalent Hsp90 inhibitors that allosterically disrupt the Hsp90-Cdc37 interaction. Here, through systematic structure-activity relationship (SAR) optimization, compound 39 (DDO-6691) with a new covalent warhead was developed, which demonstrates improved ADME properties and significantly enhanced antitumor activity. Notably, parental HCT-116 cells exhibited markedly greater sensitivity to compound 39 (IC50 > 50 μM) compared to their Cdc37-knockout counterparts. Importantly, compound 39 displayed potent tumor growth inhibition in HCT-116 xenograft mouse models. These collective findings underscore the therapeutic promise of covalent Hsp90-targeted disruption of the Hsp90-Cdc37 complex, offering a novel mechanistic approach to cancer treatment.
Collapse
Affiliation(s)
- Zi-Wen Feng
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Li Li
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- School of Pharmacy, Jiangsu Health Vocational College, Nanjing 210029, China
| | - Shi-Duo Zhang
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ying-Ji Wang
- Department of Pharmaceutical Analysis, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jia-Yue Pei
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Nan-Nan Chen
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Bei-Duo Wu
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qiu-Ling Zheng
- Department of Pharmaceutical Analysis, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qi-Dong You
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao-Ke Guo
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao-Li Xu
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
16
|
Janes KA, Lazzara MJ. Systems Biology of the Cancer Cell. Annu Rev Biomed Eng 2025; 27:1-28. [PMID: 39689262 DOI: 10.1146/annurev-bioeng-103122-030552] [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: 12/19/2024]
Abstract
Questions in cancer have engaged systems biologists for decades. During that time, the quantity of molecular data has exploded, but the need for abstractions, formal models, and simplifying insights has remained the same. This review brings together classic breakthroughs and recent findings in the field of cancer systems biology, focusing on cancer cell pathways for tumorigenesis and therapeutic response. Cancer cells mutate and transduce information from their environment to alter gene expression, metabolism, and phenotypic states. Understanding the molecular architectures that make each of these steps possible is a long-term goal of cancer systems biology pursued by iterating between quantitative models and experiments. We argue that such iteration is the best path to deploying targeted therapies intelligently so that each patient receives the maximum benefit for their cancer.
Collapse
Affiliation(s)
- Kevin A Janes
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA; ,
| | - Matthew J Lazzara
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA; ,
| |
Collapse
|
17
|
Vinogradov AE, Anatskaya OV. "Cell dedifferentiation" versus "evolutionary reversal" theories of cancer: The direct contest of transcriptomic features. Int J Cancer 2025; 156:1802-1813. [PMID: 39888036 DOI: 10.1002/ijc.35352] [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: 10/11/2024] [Revised: 01/07/2025] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
Cell dedifferentiation is considered an important hallmark of cancer. The atavistic reversal to a unicellular-like (UC) state is a less widely accepted concept, so far not included in the conventional hallmarks. The activated expression of ontogenetically earlier and evolutionary earlier genes in cancers supports both theories because ontogenesis partially recapitulates phylogenesis during cell differentiation (the cellular biogenetic law). We directly contested both types of gene signatures in stem vs. differentiated and cancer vs. normal cells, using meta-analysis of human single-cell transcriptomes (totally, 38 pairwise comparisons involving over 18,600 cells). Because compared cells can differ in proliferation rate, the correction for cell cycle activity was applied. Taken together as multiple variables in stem vs. differentiated cells analyses, the ontogenetic signature excluded the UC signature from predictive variables, usually even forcing it to change the sign of prediction (from plus to minus). In contrast, in cancer vs. normal cells, the UC signature excluded the ontogenetic signature. Thus, the direct contest decided in favor of the atavistic theory and placed a UC-like state as a central hallmark of cancer, which has a plausible evolutionarily formed mechanism (UC attractor) and can generate other hallmarks. These data suggest a paradigm shift in the understanding of oncogenesis and propose an integrative framework for cancer research. In a practical sense, the upregulation of UC signature over ontogenetic signature indicates potential tumorigenicity, which can be used in early diagnostics and regenerative medicine. For therapy, these results suggest the UC center of cellular networks as a universal target.
Collapse
Affiliation(s)
| | - Olga V Anatskaya
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
18
|
Lee Y. Principles and therapeutics of cancer. Mol Cells 2025; 48:100201. [PMID: 40024317 PMCID: PMC12034218 DOI: 10.1016/j.mocell.2025.100201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Affiliation(s)
- Yoontae Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea.
| |
Collapse
|
19
|
Liu L, Wu M, Chen Y, Cheng Y, Liu S, Zhang X, Xie Q, Cao L, Wei L, Fang Y, Jafri A, Sferra TJ, Shen A, Li L. Downregulating FGGY carbohydrate kinase domain containing promotes cell senescence by activating the p53/p21 signaling pathway in colorectal cancer. Int J Mol Med 2025; 55:81. [PMID: 40116125 PMCID: PMC11964412 DOI: 10.3892/ijmm.2025.5522] [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: 07/04/2024] [Accepted: 01/14/2025] [Indexed: 03/23/2025] Open
Abstract
Carbohydrate kinases serve an oncogenic role in several types of cancer; however, the function of FGGY carbohydrate kinase domain containing (FGGY) in colorectal cancer (CRC) remains unknown. The present study investigated the function and possible molecular mechanisms of FGGY in CRC. The results showed that elevated levels of FGGY mRNA and protein were observed in CRC tissues, and a higher expression of FGGY was associated with advanced N stage and reduced overall survival time in patients with CRC. Silencing FGGY inhibited the viability of CRC cells by inducing cell cycle arrest and promoting apoptosis in vitro, thereby attenuating tumor growth in a xenograft mouse model. FGGY knockdown also enriched the senescence‑associated heterochromatin foci (SAHF) pathway and p53 pathway, as further confirmed by enhancing senescence‑associated β‑galactosidase (SA‑β‑gal) activity, with increased levels of SAHF‑associated proteins HP1γ and trimethylation of H3K9 (H3k9me3) in CRC cells, as well as upregulation of p53 and its downstream protein p21. Furthermore, p53 knockout rescued FGGY knockdown‑mediated reductions in cell viability, SA‑β‑gal activity, and the levels of HP1γ and H3k9me3 in CRC cells. These findings indicated that FGGY could act as a newly identified potential oncogene in CRC, partially through regulating the p53/p21 signaling pathway and altering cell senescence.
Collapse
Affiliation(s)
- Liya Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Department of Scientific Research, Affiliated Sanming Integrated Medicine Hospital of Fujian University of Traditional Chinese Medicine, Sanming, Fujian 365001, P.R. China
| | - Meizhu Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Youqin Chen
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, OH 44106, USA
| | - Ying Cheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Sijia Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xinran Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Qiurong Xie
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Liujing Cao
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Lihui Wei
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Yi Fang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Anjum Jafri
- Department of Genetics and Genome Sciences, Histology Core, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Thomas J. Sferra
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, OH 44106, USA
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Li Li
- Shengli Clinical College, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Department of Health Management, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
20
|
Mossburg KJ, Barragan D, O NH, Kian AC, Maidment ADA, Cormode DP. Emerging nanoparticle-based x-ray imaging contrast agents for breast cancer screening. Nanomedicine (Lond) 2025; 20:1149-1166. [PMID: 40261216 DOI: 10.1080/17435889.2025.2496129] [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: 02/05/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025] Open
Abstract
Breast cancer is one of the most common types of cancer, however, preventive screening has contributed to a significant reduction in mortality over the past four decades. The first-line screening methods for breast cancer, such as mammography and tomosynthesis, are x-ray-based modalities. Unfortunately, their cancer detection rates are low in patients with dense breasts. These, and other high-risk women, are now encouraged to receive supplemental screening. The supplemental imaging methods are diverse, including ultrasound, MRI, nuclear imaging, and X-ray-based modalities such as breast CT and contrast-enhanced mammography/tomosynthesis. Due to their low cost and wide availability, x-ray-based modalities see significant clinical use worldwide. These techniques benefit from the use of contrast agents, which are currently iodinated small molecules designed for other purposes. Consequently, developing new contrast agents that are specifically for breast cancer screening is of interest. This review describes these modalities and the nanoparticle-based contrast agents being researched for their enhanced performance. The relevant parameters for nanoparticle-based contrast agent design are evaluated, including contrast generation and potential biointeractions. Iodinated agents are discussed for comparison. Nanoparticles covered include silver sulfide, silver telluride, gold, and bismuth sulfide-based agents, among others. Finally, perspectives on future developments in this field are offered.
Collapse
Affiliation(s)
- Katherine J Mossburg
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Diego Barragan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathaniel H O
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pharmaceutical Sciences, St. Joseph's University, Philadelphia, PA, USA
- Department of Physics, St. Joseph's University, Philadelphia, PA, USA
| | - Andrea C Kian
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew D A Maidment
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - David P Cormode
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
21
|
Skaličková M, Abramenko N, Charnavets T, Vellieux F, Leischner Fialová J, Kučnirová K, Kejík Z, Masařík M, Martásek P, Pacak K, Pacák T, Jakubek M. Interaction of Selected Anthracycline and Tetracycline Chemotherapeutics with Poly(I:C) Molecules. ACS OMEGA 2025; 10:15935-15946. [PMID: 40321536 PMCID: PMC12044458 DOI: 10.1021/acsomega.4c05483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 02/11/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025]
Abstract
Despite the natural ability of the immune system to recognize cancer and, in some patients, even to eliminate it, cancer cells have acquired numerous evading mechanisms. With the increasing knowledge and focus shifting from targeting rapidly proliferating cells with chemotherapy to modulating the immune system, there have been recent efforts to integrate (e.g., simultaneously or sequentially) various therapeutic approaches. Combining the oncolytic activity of some chemotherapeutics with immunostimulatory molecules, so-called chemoimmunotherapy, is an attractive strategy. An example of such an immunostimulatory molecule is polyinosinic:polycytidylic acid [Poly(I:C)], a synthetic analogue of double-stranded RNA characterized by rapid nuclease degradation hampering its biological activity. This study investigated the possible interactions of tetracycline and anthracycline chemotherapeutics with different commercial Poly(I:C) molecules and protection against nuclease degradation. Fluorescence spectroscopy and circular dichroism revealed an interaction of all of the selected chemotherapeutics with Poly(I:C)s and the ability of doxycycline and minocycline to prolong the resistance to RNase cleavage, respectively. The partial protection was observed in vitro as well.
Collapse
Affiliation(s)
- Markéta Skaličková
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Nikita Abramenko
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Tatsiana Charnavets
- Institute
of Biotechnology of the Czech Academy of Sciences, BIOCEV, 252
50 Vestec, Czech
Republic
| | - Frédéric Vellieux
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | | | - Kateřina Kučnirová
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Zdeněk Kejík
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Michal Masařík
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
- Department
of Physiology, Faculty of Medicine, Masaryk
University, Kamenice 5, 625 00 Brno, Czech Republic
- Department
of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625
00, Czech Republic
| | - Pavel Martásek
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Karel Pacak
- Section on
Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute
of Child Health and Human Development, National
Institutes of Health, Building 10, Room 1-3140, 10 Center Drive, Bethesda, Maryland 20892, United States
| | - Tomáš Pacák
- TumorSHOT, Italská 2581/67, Vinohrady,
Praha 2, Prague 120 00, Czech Republic
| | - Milan Jakubek
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| |
Collapse
|
22
|
Delmas D, Mialhe A, Cotte AK, Connat JL, Bouyer F, Hermetet F, Aires V. Lipid metabolism in cancer: Exploring phospholipids as potential biomarkers. Biomed Pharmacother 2025; 187:118095. [PMID: 40311223 DOI: 10.1016/j.biopha.2025.118095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/03/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Aberrant lipid metabolism is increasingly recognized as a hallmark of cancer, contributing to tumor growth, metastatic dissemination, and resistance to therapy. Cancer cells reprogram key metabolic pathways-including de novo lipogenesis, lipid uptake, and phospholipid remodeling-to sustain malignant progression and adapt to microenvironmental demands. This review summarizes current insights into the role of lipid metabolic reprogramming in oncogenesis and highlights recent advances in lipidomics that have revealed cancer type- and stage-specific lipid signatures with diagnostic and prognostic relevance. We emphasize the dual potential of lipid metabolic pathways-particularly those involving phospholipids-as sources of clinically relevant biomarkers and therapeutic targets. Enzymes and transporters involved in these pathways have emerged as promising candidates for both diagnostic applications and pharmacological intervention. We also examine persistent challenges hindering the clinical translation of lipid-based approaches, including analytical variability, insufficient biological validation, and the lack of standardized integration into clinical workflows. Furthermore, the review explores strategies to overcome these barriers, highlighting the importance of incorporating lipidomics into multi-omics frameworks, supported by advanced computational tools and AI-driven analytics, to decipher the complexity of tumor-associated metabolic networks. We discuss how such integrative approaches can facilitate the identification of actionable metabolic targets, improve the specificity and robustness of lipid-based biomarkers, and enhance patient stratification in the context of precision oncology.
Collapse
Affiliation(s)
- Dominique Delmas
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France; Centre de Lutte Contre le Cancer Georges François Leclerc Center, Dijon F-21000, France; Inserm UMS58 - Biologie Santé Dijon (BioSanD), Dijon F-21000, France.
| | - Aurélie Mialhe
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Alexia K Cotte
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Jean-Louis Connat
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Florence Bouyer
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - François Hermetet
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Virginie Aires
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| |
Collapse
|
23
|
Wang T, Weng M, Li K, Li G, Hu S, Hu Z, Li Y, Li M, Wu D, Liang Z, Yu F, Wang G, Li X. LIN28B enhances the chemosensitivity of colon cancer cells via inducing genomic instability by upsetting the balance between the production and removal of reactive oxygen species. Cancer Lett 2025; 616:217572. [PMID: 39986369 DOI: 10.1016/j.canlet.2025.217572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/08/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Genomic instability is an enabling characteristic that allows cancer cells to acquire additional hallmarks of cancer through the accumulation of alterations in driver genes. Furthermore, it creates opportunities to enhance the sensitivity of cancer cells to chemotherapeutic agents targeting DNA, owing to the presence of incomplete DNA damage repair pathways. This study identifies LIN28B as a crucial regulator of colon cancer cells' sensitivity to DNA damage- or repair-related compounds by promoting genomic instability. LIN28B mechanistically reduces glutathione (GSH) synthesis and activity by inhibiting the expression of four GSH metabolic enzymes (GCLC, G6PD, GSTM4, and GSTT2B), thereby reducing the capacity of cells to eliminate reactive oxygen species (ROS). LIN28B enhances the proinflammatory signaling pathway in cancer cells through the upregulation of ARID3A, a transcription factor that transactivates PTGES and PTGES2, resulting in increased production of PGE2, a key inflammatory mediator that can elevate ROS generation. In conclusion, LIN28B altered the equilibrium of ROS production and elimination in colon cancer, resulting in elevated ROS levels and subsequent genomic instability.
Collapse
Affiliation(s)
- Tianzhen Wang
- Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China; Department of Pathology, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, PR China
| | - Mingjiao Weng
- Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China; Department of Pathology, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, PR China; Department of Stomatology, Shenzhen Second People's Hospital, Shenzhen, 518000, PR China
| | - Kai Li
- Department of Oncology 2, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, PR China
| | - Guoli Li
- Department of Anus and Intestine Surgery, Chifeng Municipal Hospital, No.1 Middle Section of Zhaowuda Road, Chifeng, Inner Mongolia, 024000, PR China
| | - Shijie Hu
- Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China; Department of Pathology, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, PR China
| | - Ziyi Hu
- Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China; Department of Pathology, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, PR China
| | - Yanping Li
- Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China; Department of Pathology, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, PR China
| | - Muhan Li
- Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China; Department of Pathology, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, PR China
| | - Di Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, PR China
| | - Zhigang Liang
- Department of Stomatology, Shenzhen Second People's Hospital, Shenzhen, 518000, PR China.
| | - Fei Yu
- Department of Traditional Chinese Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, PR China.
| | - Guangyu Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, Heilongjiang, 150081, PR China.
| | - Xiaobo Li
- Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China; Department of Pathology, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, PR China.
| |
Collapse
|
24
|
Mohamed NM, Mohamed RH, Kennedy JF, Elhefnawi MM, Hamdy NM. A comprehensive review and in silico analysis of the role of survivin (BIRC5) in hepatocellular carcinoma hallmarks: A step toward precision. Int J Biol Macromol 2025; 311:143616. [PMID: 40306500 DOI: 10.1016/j.ijbiomac.2025.143616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/25/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Hepatocellular carcinoma (HCC) is a complex malignancy driven by the dysregulation of multiple cellular pathways. Survivin, a key member of the inhibitor of apoptosis (IAP) family, plays a central role in HCC tumorigenesis and progression. Despite significant research, a comprehensive understanding of the contributions of survivin to the hallmarks of cancer, its molecular network, and its potential as a therapeutic target remains incomplete. In this review, we integrated bioinformatics analysis with an extensive literature review to provide deeper insights into the role of survivin in HCC. Using bioinformatics tools such as the Human Protein Atlas, GEPIA, STRING, TIMER, and Metascape, we analyzed survivin expression and its functional associations and identified the top 20 coexpressed genes in HCC. These include TK1, SPC25, SGO2, PTTG1, PRR11, PLK1, NCAPH, KPNA2, KIF2C, KIF11, HJURP, GTSE1, FOXM1, CEP55, CENPA, CDCA3, CDC45, CCNB2, CCNB1 and CTD-2510F5.4. Our findings also revealed significant protein-protein interactions among these genes, which were enriched in pathways associated with the FOXM1 oncogenic signaling cascade, and biological processes such as cell cycle regulation, mitotic checkpoints, and diseases such as liver neoplasms. We also discussed the involvement of survivin in key oncogenic pathways, including the PI3K/AKT, WNT/β-catenin, Hippo, and JAK/STAT3 pathways, and its role in modulating cell cycle checkpoints, apoptosis, and autophagy. Furthermore, we explored its interactions with the tumor microenvironment, particularly its impact on immune modulation through myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages, and natural killer cell function in HCC. Additionally, we highlighted its involvement in alkylglycerone phosphate synthase (AGPS)-mediated lipid reprogramming and identified important gaps in the survivin network that warrant further investigation. This review also examined the role of survivin in cancer stemness, inflammation, and virally mediated hepatocarcinogenesis. We evaluated its potential as a diagnostic, prognostic, predictive, and pharmacodynamic biomarker in HCC, emphasizing its relevance in precision medicine. Finally, we summarized emerging survivin-targeted therapeutics and ongoing clinical trials, underscoring the need for novel strategies to effectively target survivin in HCC.
Collapse
Affiliation(s)
- Nermin M Mohamed
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt
| | - Rania Hassan Mohamed
- Department of Biochemistry, Faculty of Science, Ain Shams University, Abassia, 11566 Cairo, Egypt
| | - John F Kennedy
- Chembiotech Laboratories, Kyrewood House, Tenbury Wells, Worcestershire, United Kingdom
| | - Mahmoud M Elhefnawi
- Biomedical Informatics and Chemoinformatics Group, Informatics and Systems Department, National Research Centre, Cairo, Egypt.
| | - Nadia M Hamdy
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt.
| |
Collapse
|
25
|
Sankar S, Kalidass B, Indrakumar J, Kodiveri Muthukaliannan G. NSAID-encapsulated nanoparticles as a targeted therapeutic platform for modulating chronic inflammation and inhibiting cancer progression: a review. Inflammopharmacology 2025:10.1007/s10787-025-01760-8. [PMID: 40285986 DOI: 10.1007/s10787-025-01760-8] [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: 03/24/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025]
Abstract
Recent advancements in nanotechnology have significantly advanced nanocarrier-mediated drug delivery systems, promoting therapeutic outcomes in mitigating chronic inflammation and cancer. Nanomaterials offer significant advantages over traditional small-molecule drugs, including a high surface-area-to-volume ratio, tunable structural features, and extended bloodstream circulation time. Chronic inflammation is a well-established mechanism for malignant initiation, progression, and metastasis, promoting the potent strategy for cancer prevention and therapy. Numerous studies revealed that nonsteroidal anti-inflammatory drugs (NSAIDs) have the therapeutic ability to manage disease progression via amolerating angiogenesis and inducing apoptosis. However, prolonged intake of NSAIDs is often limited by adverse side-effects and systemic toxicities. The encapsulation of NSAIDs in a nanocarrier have materialized as a dynamic approach to mitigate the limitations by improving pharmacokinetics and pharmacodynamics, reducing off-target effects, and enhancing the drug stability. This review encompasses recent progress in the development of NSAID-based nanotherapeutics, focusing on pivotal mechanisms underlying nanoparticle-mediated drug delivery, such as improved tumor-specific targeting and strategies to overcome drug resistance. The ability of these nano-cargoes to accommodate anti-inflammatory strategies with advanced drug delivery platforms is critically evaluated. This review also highlights the transformative potential of NSAID-encapsulated nanoparticles as a multifaceted therapeutic venue for addressing chronic inflammation and mitigating cancer progression.
Collapse
Affiliation(s)
- Srivarshini Sankar
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632 014, India
| | - Bharathi Kalidass
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632 014, India
| | - Janani Indrakumar
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632 014, India
| | - Gothandam Kodiveri Muthukaliannan
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632 014, India.
| |
Collapse
|
26
|
Borlongan MC, Rodriguez T, Putthanbut N, Wang H, Lee JY. Modeling of cancer stem cells and the tumor microenvironment Via NT2/D1 cells to probe pathology and treatment for cancer and beyond. Discov Oncol 2025; 16:605. [PMID: 40272656 PMCID: PMC12022208 DOI: 10.1007/s12672-025-02158-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/17/2025] [Indexed: 04/27/2025] Open
Abstract
INTRODUCTION Unique from the other tumor cells, tumorigenic cancer stem cells (CSCs) manifest as a subpopulation of cells within the tumor that exhibit genetic and phenotypic features and signaling processes, which escape traditional anti-oncogenic treatments, thereby triggering metastases and relapses of cancers. Critical to cancer biology is the crosstalk between CSCs and tumor microenvironment (TME), implicating a CSC-based cancer immunotherapy. Cognizant of CSCs' significant role in cancer pathology and treatment, finding a biological model that recapitulates CSCs and TME may allow a better understanding of tumor onset and progression for testing CSC-based therapies. In this review paper, we examined the CSC and TME characteristics of the human embryonal carcinoma NTERA-2 clonal cell line called NTERA-2 cl.D1 or NT2/D1 cells and discussed their potential utility for research and development of treatments for cancer and central nervous system (CNS) disorders. METHODS To probe our hypotheses that NT2/D1 cells display CSC and TME properties key to tumor development, which can serve as a screening platform to test cancer and CNS therapeutics, we conducted a literature review over a 10-year period (2014-2024), focusing on PUBMED and Science Direct published articles on cellular models of cancer, with emphasis on milestone research discoveries on NT2/D1 cells relevant to CSCs and TME. We categorized the studies under pre-clinical and clinical investigations in supporting the existence of CSC and TME features in NT2/D1 cells and providing a laboratory-to-clinic translational basis for cancer and CNS therapeutics. CONCLUSIONS NT2/D1 cells stand as a feasible biological model that recapitulates the crosstalk of CSCs and TME, which may critically contribute to our understanding of cancer and CNS biology and therapeutics. Designing therapeutics against CSCs' distinct self-renewal and differentiation capacities within the TME opens new avenues for treating cancers and CNS disorders.
Collapse
Affiliation(s)
- Mia C Borlongan
- California Northstate University College of Medicine, Elk Grove, CA, 95757, USA
| | - Thomas Rodriguez
- Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Napasiri Putthanbut
- Center of Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, FL, 33612, USA
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Hongbin Wang
- California Northstate University College of Pharmacy, Elk Grove, CA, 95757, USA
| | - Jea-Young Lee
- Center of Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, FL, 33612, USA.
| |
Collapse
|
27
|
Bai L, Yi M, Xu B. Self-Assembly of Noncanonical Peptides: A New Frontier in Cancer Therapeutics and Beyond. Macromol Biosci 2025:e2500153. [PMID: 40260674 DOI: 10.1002/mabi.202500153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/03/2025] [Indexed: 04/24/2025]
Abstract
In addition to the 20 standard amino acids that form the building blocks of proteins, nature employs alternative amino acids to create specialized "noncanonical peptides." These unique peptides, found in organisms from bacteria to humans, often exhibit unconventional structures and functionalities, playing critical roles in modulating cellular processes, particularly as antibiotics. Their potential has attracted significant interest for designing novel functional materials based on noncanonical peptides. This review highlights recent advances in the generation and application of noncanonical peptide assemblies. It begins with a definition of noncanonical peptides, including classic examples that showcase their distinct structures and useful biological activities. Then the applications of noncanonical peptide assemblies in developing anticancer therapeutics are discussed, focusing on recent and representative studies that demonstrate their efficacy and versatility in targeting tumor cells. Beyond oncology, it is explored how noncanonical peptide assemblies have been utilized in biomaterials, regenerative medicine, molecular imaging and catalysis. Finally, perspectives are offered on future directions in this rapidly evolving field, emphasizing exciting opportunities and remaining challenges that will drive continued innovation in designing and applying noncanonical peptide-based assemblies.
Collapse
Affiliation(s)
- Lin Bai
- School of Chemistry, Brandeis University, 415 South St, Waltham, MA, 02453, USA
| | - Meihui Yi
- School of Chemistry, Brandeis University, 415 South St, Waltham, MA, 02453, USA
| | - Bing Xu
- School of Chemistry, Brandeis University, 415 South St, Waltham, MA, 02453, USA
| |
Collapse
|
28
|
Wu MJ, Yang SM, Fang WK, Chen TJ, Wu CY, Hsu YJ, Shen CE, Cheng YC, Hsieh WC, Yuh CH, Yang MH, Kung HJ, Wang WC. KDM4C works in concert with GATA1 to regulate heme metabolism in head and neck squamous cell carcinoma. Cell Mol Life Sci 2025; 82:170. [PMID: 40259045 PMCID: PMC12011672 DOI: 10.1007/s00018-025-05693-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 03/19/2025] [Accepted: 04/01/2025] [Indexed: 04/23/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC), the sixth most common cancer worldwide, presents significant public health challenges due to its genetic instability and late-stage diagnosis. Despite advancements in treatment, the median overall survival remains below one year, emphasizing the need for improved detection, prognosis, and therapeutic strategies. This study investigates the role of KDM4C and its interaction with GATA1 in regulating heme metabolism and tumor progression in HNSCC. KDM4C knockdown (KDM4C-KD) hindered HNSCC cell migration using in vitro assays, inhibited metastasis through zebrafish xenotransplantation, and suppressed tumor growth in mouse xenograft models. RNA-seq and CUT&Tag-seq analyses on KDM4C-KD SAS cells identified KDM4C-regulated genes, including ferrochelatase (FECH), in heme metabolism. Immunoprecipitation and docking analyses confirmed the KDM4C-GATA1 interaction. Notably, FECH overexpression in KDM4C or GATA1 knockdown cells restored cell migration, invasion, and proliferation, highlighting FECH as a crucial downstream target. KDM4 inhibitors myricetin and BPRKD022S0 (22S0) increased H3K9me3 levels, downregulated heme metabolism genes, and reduced cell survival in HNSCC cells. Zebrafish and mouse models demonstrated that these inhibitors effectively suppressed tumor growth and metastasis. Immunohistochemical analysis of HNSCC patient samples revealed high KDM4C and GATA1 expression correlated with advanced clinical stages and poor survival outcomes. Our findings elucidate the critical role of the KDM4C/GATA1-FECH axis in HNSCC progression and suggest that targeting this pathway with KDM4 inhibitors shows promising therapeutic potential for HNSCC treatment.
Collapse
Affiliation(s)
- Meng-Jen Wu
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Shan-Min Yang
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Wei-Kai Fang
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Tsan-Jan Chen
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Chun-Yi Wu
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Yen-Jung Hsu
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Cheng-En Shen
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Yu-Chia Cheng
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Wan-Chen Hsieh
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Chiou-Hwa Yuh
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, 35053, Taiwan, ROC
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan, ROC
| | - Hsing-Jien Kung
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, 11031, Taiwan, ROC
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, University of California Davis Cancer Centre, Sacramento, CA, 95817, USA
| | - Wen-Ching Wang
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC.
| |
Collapse
|
29
|
Zhang Y, Ye L, Qin Y, Qiu C, Sun Q, Fan T, Chen Y, Jiang Y. Serum metabolomics to identify molecular subtypes and predict XELOX efficacy in colorectal cancer. Sci Rep 2025; 15:13671. [PMID: 40258977 PMCID: PMC12012017 DOI: 10.1038/s41598-025-97463-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/04/2025] [Indexed: 04/23/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers; however, accurately predicting prognosis based on existing molecular subtypes remains challenging. The XELOX regimen, which combines oxaliplatin and capecitabine, is the cornerstone of chemotherapy for CRC treatment. However, there is a notable lack of reliable predictive models for determining the sensitivity of this treatment. This study aimed to establish a novel classification system for CRC and develop a predictive model for XELOX chemotherapeutic sensitivity using serum metabolomics. We recruited 89 patients with CRC and 89 age- and sex-matched healthy controls for untargeted metabolomic studies to identify tumor-specific serum metabolites. The patients were grouped into distinct metabolic subtypes using unsupervised clustering. A serum metabolite combination predictive of the efficacy of XELOX was established using Cox regression analysis in 34 patients with stage III CRC. Using unsupervised clustering based on the serum metabolites, three distinct clusters were identified. Notably, Cluster 3, which was characterized by uniform lipid and amino acid levels, demonstrated the best prognosis. Our analysis revealed that D-glucose 6-phosphate, presqualene diphosphate, and leukotriene B4 levels were negatively correlated with XELOX sensitivity, whereas 15-HETE and N-acetyl-l-methionine levels were positively correlated. Based on these findings, we constructed a predictive model validated in an independent cohort of 34 patients with stage III CRC. In summary, this study identified a novel classification of CRC based on serum metabolites and developed a potential prognostic model for XELOX chemotherapeutic efficacy, which may have direct effects on the treatment and prognosis of CRC.
Collapse
Affiliation(s)
- Yijie Zhang
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Lizhen Ye
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China
| | - Ying Qin
- Department of Gastrointestinal Surgery, Shenzhen Second People'S Hospital, Shenzhen, China
| | - Cheng Qiu
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China
| | - Qinsheng Sun
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Tingting Fan
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yan Chen
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China.
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China.
| | - Yuyang Jiang
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
30
|
Duzgun D, Oltean S. Aberrant Splicing as a Mechanism for Resistance to Cancer Therapies. Cancers (Basel) 2025; 17:1381. [PMID: 40282556 PMCID: PMC12025770 DOI: 10.3390/cancers17081381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Cancer is biologically diverse, highly heterogeneous, and associated with molecular alterations, significantly contributing to mortality worldwide. Currently, cancer patients are subjected to single or combination treatments comprising chemotherapy, surgery, immunotherapy, radiation therapy, and targeted therapy. Chemotherapy remains the first line of treatment in cancer but faces a major obstacle in the form of chemoresistance. This obstacle has resulted in relapses and poor patient survival due to decreased treatment efficacy. Aberrant pre-mRNA alternative splicing can significantly modulate gene expression and function involved in the resistance mechanisms, potentially shaping the intricate landscape of tumour chemoresistance. Thus, novel strategies targeting abnormal pre-mRNA alternative splicing and understanding the molecular mechanisms of chemotherapy resistance could aid in overcoming the chemotherapeutic challenges. This review first highlights drug targets, drug pumps, detoxification mechanisms, DNA damage response, and evasion of apoptosis and cell death as key molecular mechanisms involved in chemotherapy resistance. Furthermore, the review discusses the progress of research on the dysregulation of alternative splicing and molecular targets involved in chemotherapy resistance in major cancer types.
Collapse
Affiliation(s)
| | - Sebastian Oltean
- Department of Clinical and Biomedical Sciences, Faculty of Health Sciences, University of Exeter, Exeter EX1 2LU, UK
| |
Collapse
|
31
|
Perini F, Di Franco C, Briganti A. Dysmagnesemia Incidence in Hospitalized Dogs and Cats: A Retrospective Study. Animals (Basel) 2025; 15:1169. [PMID: 40282004 PMCID: PMC12024414 DOI: 10.3390/ani15081169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
Magnesium plays a vital role in the body. This retrospective study aimed to evaluate dysmagnesemia incidence in hospitalized patients. Medical records of 430 dogs and 310 cats were reviewed, including patients with at least one venous blood gas analysis upon admission. Normal ionized magnesium values were considered 0.5-1 mmol/L for both species, according to the machine range. Data collected included patient demographics, hospitalization details, and outcome. In dogs, hypomagnesemia occurred in 35.5%, hypermagnesemia in 1.1%, and normomagnesemia in 62.2%. No survival differences were observed, but males showed a higher hypomagnesemia incidence. Neurological (51%), neoplastic (50%), and endocrine (42%) diseases were most associated with hypomagnesemia. In cats, hypomagnesemia was found in 6.8%, hypermagnesemia in 8%, and normomagnesemia in 85.2%. Hypermagnesemic cats had 2.3 times higher mortality. Endocrine (28.6%), systemic (13.6%), and urinary (12.9%) disorders had a higher incidence of hypermagnesemia. Dysmagnesemia was not linked to hospitalization length or blood pressure changes. In conclusion, dogs showed a high incidence of hypomagnesemia that was not associated with increased mortality. In contrast, although hypermagnesemia had a low incidence in cats, it was associated with increased mortality.
Collapse
Affiliation(s)
| | - Chiara Di Franco
- Department of Veterinary Science, University of Pisa, 56124 Pisa, Italy; (F.P.); (A.B.)
| | | |
Collapse
|
32
|
Han Z, Wen L. G-quadruplex in cancer energy metabolism: A potential therapeutic target. Biochim Biophys Acta Gen Subj 2025; 1869:130810. [PMID: 40254103 DOI: 10.1016/j.bbagen.2025.130810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
In recent years, energy metabolism in cancer has received increasing attention as an important component of tumor biology, and the functions of transcription factors, mitochondria, reactive oxygen species (ROS) and the autophagy-lysosome system in which have been elucidated. G-quadruplex (G4) is a molecular switch that regulates gene transcription or translation. As an anticancer target, the effect of G4 on cancer cell proliferation, apoptosis, cycle and autophagy has been recognized. The energy metabolism system is a unified whole composed of transcription factors, metabolic regulators, metabolites and signaling pathways that run through the entire cancer process. However, the role of G4 in this complex metabolic network has not been systematically elucidated. In this review, we analyze the close correlation between G4 and transcription factors, mitochondria, ROS and the autophagy-lysosome system and suggest that G4 can exert a marked effect on cancer energy metabolism by regulating the above mentioned key regulatory elements. The anticancer effects of some G4 ligands through regulation of energy metabolism have also been summarized, confirming the clear involvement of G4 in energy metabolism. Although much more research is needed, we propose that G4 may play a critical role in the complex energy metabolism system of cancer, which is a promising target for anticancer strategies focusing on energy metabolism.
Collapse
Affiliation(s)
- Zongqiang Han
- Department of Laboratory Medicine, Beijing Xiaotangshan Hospital, Beijing 102211, China
| | - Lina Wen
- Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China.
| |
Collapse
|
33
|
Luo Z, Wang Y, Zeng S, Yu L, Zhao Y, Wang H, Fan Y, Zhang Y, Wang L, Li Y, Niu Z, Zhang X, Zhang Y. Harnessing lysosomal genetics: development of a risk stratification panel and unveiling of DPP7 as a biomarker for colon adenocarcinoma. J Genet Genomics 2025:S1673-8527(25)00118-3. [PMID: 40254156 DOI: 10.1016/j.jgg.2025.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 04/08/2025] [Accepted: 04/12/2025] [Indexed: 04/22/2025]
Abstract
Lysosomal dysfunction has been implicated in the progression of colon adenocarcinoma (COAD), yet the prognostic significance and therapeutic potential of lysosome-related genes (LRGs) remain underexplored. In this study, we construct a 6-LRG-based prognostic risk stratification model (DPP7, ADAM8, CD1B, LRP2, ATP6V1C2, and PLAAT3) by integrating LASSO and Cox regression analyses. Stratifying patients based on median risk scores, we demonstrate that high-risk patients exhibit significantly worse clinical outcomes across the TCGA cohort and five independent GEO datasets. Furthermore, this panel outperforms 136 previously published models in terms of predictive accuracy for 1-, 3-, and 5-year survival rates. Validation multiplex immunofluorescence using an in-house tissue microarray cohort confirms the 6-LRG signature serves as an independent prognostic factor. Additionally, high-risk patients exhibit distinct immunosuppressive tumor microenvironment and aggressive malignancy characteristics. Functional depletion of DPP7 significantly inhibits tumor cell proliferation, migration, and metastasis in both in vitro and in vivo settings. Moreover, DPP7 silencing attenuates epithelial-mesenchymal transition, as evidenced by the upregulation of E-cadherin and downregulation of N-cadherin, Vimentin, and Snail. In conclusion, this study establishes an LRG-based model for COAD prognostic prediction and nominates DPP7 as a promising therapeutic target for COAD treatment.
Collapse
Affiliation(s)
- Zhengdong Luo
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Yanlei Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Shunjie Zeng
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Longchen Yu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Yuxiao Zhao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Hong Wang
- Department of Anesthesiology, Yidu Central Hospital, Weifang Medical University, Qingzhou, Shandong 262500, China
| | - Yingjing Fan
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Yanli Zhang
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Jinan, Shandong 250000, China
| | - Lili Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Yaping Li
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Zhongfang Niu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Xin Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China.
| | - Yi Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China.
| |
Collapse
|
34
|
Jaiswal N, Mahata N, Chanda N. Nanogold-albumin conjugates: transformative approaches for next-generation cancer therapy and diagnostics. NANOSCALE 2025. [PMID: 40237258 DOI: 10.1039/d4nr05279j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Nanogold-albumin conjugates have garnered significant attention as a highly adaptable theranostic platform, capable of delivering a wide range of therapeutics, from small-molecule drugs to larger biomolecules, while offering promising applications for monitoring and managing cancer. The remarkable theranostic capabilities of these conjugates stem from the combined strengths of gold and albumin, which provide low toxicity, a large surface area, customizable surface chemistry, and unique optical properties, all contributing to their potential in cancer therapy. This review delves into the design and development of two primary types of nanogold-albumin conjugate: supramolecular albumin-coated gold nanoparticles (GNP-BSA/HSA) and albumin-templated ultra-small gold nanoclusters (GNC-BSA/HSA). Each strategy offers distinct advantages, enabling the fine-tuning of conjugate properties to optimize therapeutic delivery and facilitate cancer-specific bio-sensing. The integration of gold and albumin further improves biocompatibility, extends circulation time, and enhances tumor targeting, making these conjugates an attractive option for cancer treatment. The review also focuses on the refinement of surface chemistry to achieve precise targeting of cancer cells, as well as the challenges and future prospects for advancing nanogold-albumin systems in clinical applications.
Collapse
Affiliation(s)
- Namita Jaiswal
- Human Centered Robotics and Cybernetics Group, CSIR-Central Mechanical Engineering Research Institute, Durgapur, India.
- Department of Biotechnology, National Institute of Technology (NIT), Durgapur, India
| | - Nibedita Mahata
- Department of Biotechnology, National Institute of Technology (NIT), Durgapur, India
| | - Nripen Chanda
- Human Centered Robotics and Cybernetics Group, CSIR-Central Mechanical Engineering Research Institute, Durgapur, India.
| |
Collapse
|
35
|
Tang R, Luo S, Liu H, Sun Y, Liu M, Li L, Ren H, Angele MK, Börner N, Yu K, Guo Z, Yin G, Luo H. Circulating Tumor Microenvironment in Metastasis. Cancer Res 2025; 85:1354-1367. [PMID: 39992721 PMCID: PMC11997552 DOI: 10.1158/0008-5472.can-24-1241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 11/12/2024] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Activation of invasion and metastasis is a central hallmark of cancer, contributing to the primary cause of death for patients with cancer. In the multistep metastatic process, cancer cells must infiltrate the circulation, survive, arrest at capillary beds, extravasate, and form metastatic clones in distant organs. However, only a small proportion of circulating tumor cells (CTC) successfully form metastases, with transit of CTCs in the circulation being the rate-limiting step. The fate of CTCs is influenced by the circulating tumor microenvironment (cTME), which encompasses factors affecting their biological behaviors in the circulation. This liquid and flowing microenvironment differs significantly from the primary TME or the premetastatic niche. This review summarizes the latest advancements in identifying the biophysical cues, key components, and biological roles of the cTME, highlighting the network among biophysical attributes, blood cells, and nonblood factors in cancer metastasis. In addition to the potential of the cTME as a therapeutic target for inhibiting metastasis, the cTME could also represent as a biomarker for predicting patient outcomes and developing strategies for treating cancer.
Collapse
Affiliation(s)
- Rui Tang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Shujuan Luo
- Department of Obstetrics, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Liu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yan Sun
- Department of Cell Biology and Medical Genetics, Basic Medical School, Chongqing Medical University, Chongqing, China
| | - Manran Liu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Lu Li
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Haoyu Ren
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Martin K. Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich Munich, Germany
| | - Nikolaus Börner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich Munich, Germany
| | - Keda Yu
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Zufeng Guo
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Guobing Yin
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Haojun Luo
- Department of Thyroid and Breast Surgery, Renji Hospital, School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
36
|
Zhu X, Li Z, Chen L, Li L, Ouyang M, Zhou H, Xiao K, Lin L, Chu PK, Zhou C, Xun C, Yang L, Huang W, Ding X. Exosomes delivering miR-129-5p combined with sorafenib ameliorate hepatocellular carcinoma progression via the KCTD1/HIF-1α/VEGF pathway. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01044-x. [PMID: 40227531 DOI: 10.1007/s13402-025-01044-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Potassium channel tetramerization domain-containing 1 (KCTD1) plays a critical role in transcriptional regulation and adipogenesis, but its significance in hepatocellular cancer (HCC) has not been reported. METHODS Immunohistochemistry, Western blotting and quantitative real-time PCR analysis were performed to assess the expression of KCTD1 and related genes in HCC cells. MTT assays, colony formation, cell migration, invasion and the in-vivo mouse models were utilized to evaluate the function of KCTD1 in HCC progression. Co-immunoprecipitation, chromatin immunoprecipitation and luciferase reporter assays were conducted to elucidate the molecular mechanisms of KCTD1 in HCC. RESULTS KCTD1 expression was increased in human HCC tissues and closely associated with advanced tumor stages. KCTD1 overexpression enhanced growth, migration, and invasion of Huh7 and HepG2 cells both in vitro and in vivo, while KCTD1 knockdown reversed these effects in MHCC97H cells. Mechanistically, KCTD1 interacted with hypoxia-inducible factor 1 alpha (HIF-1α) and enhanced HIF-1α protein stability with the inhibited prolyl-hydroxylases (PHD)/Von Hippel-Lindau (VHL) pathway, consequently activating the Vascular Endothelial Growth Factor (VEGF)/VEGFR2 pathway in HCC cells. Sorafenib and KCTD1 knockdown synergistically inhibited intrahepatic tumor growth following in situ injection of MHCC97H cells. miR-129-5p downregulated KCTD1 by binding to KCTD1 3'UTR. Finally, 45 µg exosomes from miR-129-5p-overexpressing MHCC97H cells combined with 25 mg/kg sorafenib to decrease HCC tumor size. CONCLUSIONS These results suggested that KCTD1 protects HIF-1α from degradation and activates the VEGF signaling cascade to enhance HCC progression. Therefore, KCTD1 may serve as a novel target of HCC and pave the way for an efficient combined therapy in advanced HCC.
Collapse
Affiliation(s)
- Xinyu Zhu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Zhiwei Li
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Li Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Limin Li
- College of Engineering and Design, Hunan Normal University, Taozihu Road No. 68, Changsha, 410081, China.
| | - Mi Ouyang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Hao Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Kai Xiao
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Ling Lin
- Department of Hepatobiliary and Pancreatic Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Paul K Chu
- Department of Physics, Department of Materials Science and Engineering, and Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Chang Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Chengfeng Xun
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Liu Yang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Wenhuan Huang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Xiaofeng Ding
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China.
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China.
| |
Collapse
|
37
|
Chornenkyy Y, Vyas M, Deshpande V. The future is now: advancing p53 immunohistochemistry in Barrett's oesophagus and its implication for the everyday pathologist. Histopathology 2025. [PMID: 40223170 DOI: 10.1111/his.15442] [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
Barrett's oesophagus (BE) is a precancerous condition where the normal squamous epithelium of the distal oesophagus is replaced by specialised intestinal-type columnar epithelium. Patients with BE have an increased risk of developing oesophageal adenocarcinoma (EAC). p53 is a tumour suppressor protein frequently mutated in BE, and its overexpression is detectable by immunohistochemistry (IHC) and correlates with TP53 mutations. This review summarises recent literature on p53 IHC as a diagnostic aid for dysplasia and a predictive biomarker of neoplastic progression risk in BE. While there is a vast amount of literature on this topic, there are no established criteria for what constitutes an abnormal p53 immunohistochemical result in the setting of BE. Multiple studies show that p53 IHC improves interobserver agreement and diagnostic confidence for low-grade dysplasia, high-grade dysplasia and EAC compared to histology alone. Establishing easy-to-use, reproducible and practical diagnostic criteria that can be applied to routine daily practice is urgently needed. p53 IHC overexpression in non-dysplastic BE indicates an increased neoplastic progression risk. Emerging technologies such as next-generation sequencing may offer higher sensitivity for detecting neoplastic clones. While the haematoxylin and eosin stain remains the most powerful tool, p53 IHC is a valuable adjunct for determining and diagnosing dysplasia. Although p53 is helpful in predicting the risk of dysplasia in non-dysplastic Barrett's oesophagus, its sensitivity is low, limiting its routine use in this context.
Collapse
Affiliation(s)
- Yevgen Chornenkyy
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Monika Vyas
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Vikram Deshpande
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
38
|
Saadh MJ, Bishoyi AK, Ballal S, Singh A, Kareem RA, Devi A, Sharma GC, Naidu KS, Sead FF. MicroRNAs as behind-the-scenes molecules in breast cancer metastasis and their therapeutic role through novel microRNA-based delivery strategies. Gene 2025; 944:149272. [PMID: 39894085 DOI: 10.1016/j.gene.2025.149272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Breast cancer is the primary cause of cancer-related death and the most frequent malignancy among women in Western countries. Although there have been advancements in combination treatments and targeted therapies for the metastatic diseases management, metastatic breast cancer is still the second most common cause of cancer-related deaths among U.S. women. The routes of metastasis encompass invasion, intravasation, circulation, extravasation, infiltration into a remote location to establish a metastatic niche, and the formation of micro-metastases in a new environment. Each of these processes is regulated by changes in gene expression. MicroRNAs (miRNAs) are widely expressed by a variety of organisms and have a key role in cell activities including suppressing or promoting cancer through regulating various pathways. Target gene expression is post-transcriptionally regulated by miRNAs, which contribute to the development, spread, and metastasis of breast cancer. In this study, we comprehensively discussed the role of miRNAs as predictors of breast cancer metastasis, their correlation with the spread of the disease to certain organs, and their potential application as targets for breast cancer treatment. We also provided molecular mechanisms of miRNAs in the progression of breast cancer, as well as current challenges in miRNA-based therapeutic approaches. Furthermore, as one of the primary issues with the treatment of solid malignancies is the efficient delivery of miRNAs, we examined a number of cutting-edge carriers for miRNA-based therapies and CRISPR/Cas9 as a targeted therapy for breast cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India
| | | | - Anita Devi
- Department of Chemistry Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Fadhil Faez Sead
- Department of Dentistry, College of Dentistry, The Islamic University, Najaf, Iraq; Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| |
Collapse
|
39
|
Simpson KL, Rothwell DG, Blackhall F, Dive C. Challenges of small cell lung cancer heterogeneity and phenotypic plasticity. Nat Rev Cancer 2025:10.1038/s41568-025-00803-0. [PMID: 40211072 DOI: 10.1038/s41568-025-00803-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 04/12/2025]
Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine malignancy with ~7% 5-year overall survival reflecting early metastasis and rapid acquired chemoresistance. Immunotherapy briefly extends overall survival in ~15% cases, yet predictive biomarkers are lacking. Targeted therapies are beginning to show promise, with a recently approved delta-like ligand 3 (DLL3)-targeted therapy impacting the treatment landscape. The increased availability of patient-faithful models, accumulating human tumour biobanks and numerous comprehensive molecular profiling studies have collectively facilitated the mapping and understanding of substantial intertumoural and intratumoural heterogeneity. Beyond the almost ubiquitous loss of wild-type p53 and RB1, SCLC is characterized by heterogeneously mis-regulated expression of MYC family members, yes-associated protein 1 (YAP1), NOTCH pathway signalling, anti-apoptotic BCL2 and epigenetic regulators. Molecular subtypes are based on the neurogenic transcription factors achaete-scute homologue 1 (ASCL1) and neurogenic differentiation factor 1 (NEUROD1), the rarer non-neuroendocrine transcription factor POU class 2 homeobox 3 (POU2F3), and immune- and inflammation-related signatures. Furthermore, SCLC shows phenotypic plasticity, including neuroendocrine-to-non-neuroendocrine transition driven by NOTCH signalling, which is associated with disease progression, chemoresistance and immune modulation and, in mouse models, with metastasis. Although these features pose substantial challenges, understanding the molecular vulnerabilities of transcription factor subtypes, the functional relevance of plasticity and cell cooperation offer opportunities for personalized therapies informed by liquid and tissue biomarkers.
Collapse
Affiliation(s)
- Kathryn L Simpson
- SCLC Biology Group, Cancer Research UK Manchester Institute, Manchester, UK
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
| | - Dominic G Rothwell
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
| | - Fiona Blackhall
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Medical Oncology, Christie Hospital National Health Service, Foundation Trust, Manchester, UK
| | - Caroline Dive
- SCLC Biology Group, Cancer Research UK Manchester Institute, Manchester, UK.
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK.
- CRUK Lung Cancer Centre of Excellence, Manchester, UK.
| |
Collapse
|
40
|
Croce CM, Vaux D, Strasser A, Opferman JT, Czabotar PE, Fesik SW. The BCL-2 protein family: from discovery to drug development. Cell Death Differ 2025:10.1038/s41418-025-01481-z. [PMID: 40204952 DOI: 10.1038/s41418-025-01481-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/24/2025] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
The landmark discovery of the BCL-2 gene and then its function marked the identification of inhibition of apoptotic cell death as a crucial novel mechanism driving cancer development and launched the quest to discover the molecular control of apoptosis. This work culminated in the generation of specific inhibitors that are now in clinical use, saving and improving tens of thousands of lives annually. Here, some of the original players of this story, describe the sequence of critical discoveries. The t(14;18) chromosomal translocation, frequently observed in follicular lymphoma, allowed the identification and the cloning of a novel oncogene (BCL-2) juxtaposed to the immunoglobulin heavy chain gene locus (IgH). Of note, BCL-2 acted in a distinct manner as compared to then already known oncogenic proteins like ABL and c-MYC. BCL-2 did not promote cell proliferation but inhibited cell death, as originally shown in growth factor dependent haematopoietic progenitor cell lines (e.g., FDC-P1) and in Eμ-Myc/Eμ-Bcl-2 double transgenic mice. Following a rapid expansion of the BCL-2 protein family, the Abbott Laboratories solved the first structure of BCL-XL and subsequently the BCL-XL/BAK peptide complex, opening the way to understanding the structures of other BCL-2 family members and, finally, to the generation of inhibitors of the different pro-survival BCL-2 proteins, thanks to the efforts of Servier/Norvartis, Genentech/WEHI, AbbVie, Amgen, Prelude and Gilead. Although the BCL-2 inhibitor Venetoclax is in clinical use and inhibitors of BCL-XL and MCL-1 are undergoing clinical trials, several questions remain on whether therapeutic windows can be achieved and what other agents should be used in combination with BH3 mimetics to achieve optimal therapeutic impact for cancer therapy. Finally, the control of the expression of BH3-only proteins and pro-survival BCL-2 family members needs to be better understood as this may identify novel targets for cancer therapy. This story is still not concluded!
Collapse
Affiliation(s)
- Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| | - David Vaux
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| | - Andreas Strasser
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| | - Joseph T Opferman
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Peter E Czabotar
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| | - Stephen W Fesik
- Department of Biochemistry, Pharmacology and Chemistry, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
41
|
Tillman L, Margalef Rieres J, Ahjem E, Bishop-Guest F, McGrath M, Hatrick H, Pranjol MZI. Thinking Outside the Therapeutic Box: The Potential of Polyphenols in Preventing Chemotherapy-Induced Endothelial Dysfunction. Cells 2025; 14:566. [PMID: 40277892 PMCID: PMC12026109 DOI: 10.3390/cells14080566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
The numerous side effects and adverse health implications associated with chemotherapies have long plagued the field of cancer care. Whilst in some cases a curative measure, this highly toxic intervention consistently scores poorly on quantitative measures of tolerability and safety. Of these side effects, cardiac and microvascular defects pose the greatest health risk and are the leading cause of death amongst cancer survivors who do not succumb to relapse. In fact, in many low-grade cancers, the risk of recurrence is far outweighed by the cardiovascular risk of morbidity. As such, there is a pressing need to improve outcomes within these populations. Polyphenols are a group of naturally occurring metabolites that have shown potential vasoprotective effects. Studies suggest they possess antioxidant and anti-inflammatory activities, in addition to directly modulating vascular signalling pathways and gene expression. Leveraging these properties may help counteract the vascular toxicity induced by chemotherapy. In this review, we outline the main mechanisms by which the endothelium is damaged by chemotherapeutic agents and discuss the ability of polyphenols to counteract such side effects. We suggest future considerations that may help overcome some of the published limitations of these compounds that have stalled their clinical success. Finally, we briefly explore their pharmacological properties and how novel approaches could enhance their efficacy while minimising treatment-related side effects.
Collapse
Affiliation(s)
- Luke Tillman
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (L.T.); (J.M.R.); (M.M.); (H.H.)
| | - Jaume Margalef Rieres
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (L.T.); (J.M.R.); (M.M.); (H.H.)
| | - Elena Ahjem
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (L.T.); (J.M.R.); (M.M.); (H.H.)
| | - Fynn Bishop-Guest
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (L.T.); (J.M.R.); (M.M.); (H.H.)
| | - Meghan McGrath
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (L.T.); (J.M.R.); (M.M.); (H.H.)
| | - Helena Hatrick
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (L.T.); (J.M.R.); (M.M.); (H.H.)
| | | |
Collapse
|
42
|
Singh K, Jacobs BA. A Network Based Model for Predicting Spatial Progression of Metastasis. Bull Math Biol 2025; 87:65. [PMID: 40202589 PMCID: PMC11982130 DOI: 10.1007/s11538-025-01441-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 03/10/2025] [Indexed: 04/10/2025]
Abstract
Metastatic cancer is reported to have a mortality rate of 90%. Understanding the underlying principles of metastasis and quantifying them through mathematical modelling provides insights into potential treatment regimes. This work presents a partial differential equation based mathematical model embedded on a network, representing the organs and the blood vessels between them, with the aim of predicting likely secondary metastatic sites. Through this framework the relationship between metastasis and blood flow and between metastasis and the diffusive behaviour of cancer is explored. An analysis of the model predictions showed a good correlation with clinical data for some cancer types, particularly for cancers originating in the gut and liver. The model also predicts an inverse relationship between blood velocity and the concentration of cancer cells in secondary organs. Finally, for anisotropic diffusive behaviour, where the cancer experiences greater diffusivity in one direction, metastatic efficiency decreased. This is aligned with the clinical observation that gliomas of the brain, which typically show anisotropic diffusive behaviour, exhibit fewer metastases. The investigation yields some valuable results for clinical practitioners and researchers-as it clarifies some aspects of cancer that have hitherto been difficult to study, such as the impact of differing diffusive behaviours and blood flow rates on the global spread of cancer. The model provides a good framework for studying cancer progression using cancer-specific information when simulating metastasis.
Collapse
Affiliation(s)
- Khimeer Singh
- School of Computational and Applied Mathematics, University of the Witwatersrand, 1 Jan Smuts Avenue, Johannesburg, 2017, Gauteng, South Africa.
| | - Byron A Jacobs
- Department of Mathematics and Applied Mathematics, University of Johannesburg, Auckland Park, PO Box 524, Johannesburg, 2006, Gauteng, South Africa
| |
Collapse
|
43
|
Yuan D, Ma R, Ye H, Liu W. A prediction model for predicting relapsed-free survival of early-stage invasive breast cancer patients with hormone receptor positive based on Ki67, HER2 and TOP2A. Front Oncol 2025; 15:1552937. [PMID: 40270612 PMCID: PMC12014635 DOI: 10.3389/fonc.2025.1552937] [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: 12/29/2024] [Accepted: 03/24/2025] [Indexed: 04/25/2025] Open
Abstract
Objective The purpose of the current study was to determine the relationship between ribonucleotide reductase M1 (RRM1), topoisomerase II alpha (TOP2A), Thymidylate synthase (TYMS), class III beta-tubulin (TUBB3) and phosphatase and tensin homolog (PTEN) expressions and relapse-free survival (RFS) in early-stage invasive breast cancer (IBC) patients with hormone receptor positive (HR+), as well as to develop a nomogram model for forecasting RFS. Methods Early-stage IBC patients with HR+ who were diagnosed and treated at the First Affiliated Hospital of Xi'an Jiaotong University from June 2017 to December 2020 were enrolled in this study. The survival analysis was performed by utilizing the Kaplan-Meier method, and the risk factors linked to patient RFS were determined by performing Cox regression analysis. The nomogram for predicting RFS in early-stage IBC patients with HR+ was stablished and validated based on the results of the Cox regression analysis. Results In total, 126 early-stage IBC patients with HR+ were included in the current study. Among these patients, 23 cases experienced relapse after surgery, with a median RFS of 29 months. Significant relationships were observed between TYMS, RRM1, TUBB3, TOP2 and PTEN, Ki67 and human epidermal growth factor receptor 2 (HER2) and patient RFS. Cox regression analysis revealed that chemotherapy and higher expression levels of TOP2A, HER2 and Ki67 were independent predictors of RFS in early-stage IBC patients with HR+. The nomogram we constructed using the above independent risk factors exhibited good ability for predicting RFS in early-stage IBC patients with HR+. Conclusion Chemotherapy, TOP2A, HER2, and Ki67 expression were independent predictors of RFS in early-stage IBC patients with HR+. The nomogram we developed using these predictors is a reliable tool for predicting RFS in this patient population.
Collapse
Affiliation(s)
- Dawei Yuan
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Rulan Ma
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Haixia Ye
- The Second Clinical College, Department of Medicine, Wuhan University, Wuhan, China
| | - Wenbo Liu
- Department of Plastic and Cosmetic Maxillofacial Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shannxi, China
| |
Collapse
|
44
|
Vidak E, Vizovišek M, Kavčič N, Biasizzo M, Fonović M, Turk B. Apoptotic Caspases-3 and -7 Cleave Extracellular Domains of Membrane-Bound Proteins from MDA-MB-231 Breast Cancer Cells. Int J Mol Sci 2025; 26:3466. [PMID: 40331965 DOI: 10.3390/ijms26083466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
Apoptotic executioner caspases-3 and -7 are the main proteases responsible for the execution of apoptosis. Apoptosis is the main form of programmed cell death involved in organism development and maintenance of homeostasis and is commonly impaired in various pathologies. Predominately an immunologically silent form of cell death, it can become immunogenic upon loss of membrane integrity during progression to secondary necrosis, which mostly occurs when apoptotic bodies are not efficiently cleared by efferocytosis. In cancer, the efferocytic capacity can be overwhelmed following chemotherapeutic treatment, thereby providing an opportunity for the potential extracellular functions of executioner apoptotic caspases in the tumor microenvironment. By triggering apoptosis in Jurkat E6.1 acute T cell leukemia cells, we demonstrated that during progression to secondary necrosis, executioner caspases-3 and -7 can be found in the extracellular space. Furthermore, we showed that extracellularly active caspases-3 and -7 can cleave extracellular domains of membrane-bound proteins from MDA-MB-231 breast cancer cells, a function generally executed in the tumor microenvironment by several extracellular proteases from metalloprotease and cathepsin families. As such, this study provides the evidence for the potential involvement of apoptotic caspases-3 and -7 in extracellular proteolytic networks. Presented mass spectrometry data are available via ProteomeXchange with identifier PXD061399.
Collapse
Affiliation(s)
- Eva Vidak
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
- Jožef Stefan International Postgraduate School, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
| | - Matej Vizovišek
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Nežka Kavčič
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Monika Biasizzo
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Marko Fonović
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
45
|
Lu M, Ren Y, Feng S, Wang S, Xia W, Gu B, Shen Y, Yue A, Li N, Zhang Y, Zhong J. MDM2 inhibitor induces apoptosis in colon cancer cells through activation of the CHOP-DR5 pathway, independent of p53 phenotype. Front Pharmacol 2025; 16:1508421. [PMID: 40264676 PMCID: PMC12011796 DOI: 10.3389/fphar.2025.1508421] [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: 10/09/2024] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction Murine double minute 2 (MDM2), a key negative regulator of p53, forms a feedback loop with p53 to drive tumor progression, including colorectal cancer. Nutlin-3a, an MDM2 inhibitor, induces apoptosis in wild-type p53 tumors, but its effects on p53-mutated cancers and potential p53-independent apoptotic mechanisms remain unclear. Methods We investigated Nutlin-3a's effects on colon cancer cells with varying p53 phenotypes. Endoplasmic reticulum (ER) stress-associated CHOP was detected and knocked down to explore mechanisms. In vitro and in vivo experiments assessed Nutlin-3a's synergy with 5-fluorouracil and TRAIL. Results Nutlin-3a activated caspase-8-dependent extrinsic apoptosis in colon cancer cells via DR5 upregulation, independent of p53 status. ER stress and CHOP activation mediated DR5 induction, driven by calcium release. Combined Nutlin-3a treatment enhanced sensitivity to 5-fluorouracil and TRAIL in vitro and in vivo through caspase-8 pathway activation. Discussion These findings reveal a novel p53-independent apoptotic mechanism of Nutlin-3a involving ER stress and death receptor signaling. This pathway highlights Nutlin-3a's potential as an adjuvant therapy for colon cancer, even in p53-mutated tumors, by enhancing chemotherapeutic efficacy through extrinsic apoptosis.
Collapse
Affiliation(s)
- Manman Lu
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yingli Ren
- College of Medicine, Henan Polytechnic University, Jiaozuo, China
| | - Sijia Feng
- Henan Province Engineering Technology Research Center of Tumor Diagnostic Biomarkers and RNA Interference Drugs, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Shenggen Wang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Weiyue Xia
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Baoru Gu
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yuhou Shen
- Department of Abdominal Surgical Oncology Ward 2, Xinxiang Central Hospital, Xinxiang, China
- Department of Abdominal Surgical Oncology Ward 2, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, China
| | - Aimin Yue
- Department of Abdominal Surgical Oncology Ward 2, Xinxiang Central Hospital, Xinxiang, China
- Department of Abdominal Surgical Oncology Ward 2, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, China
| | - Na Li
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yongxi Zhang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Province Engineering Technology Research Center of Tumor Diagnostic Biomarkers and RNA Interference Drugs, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiateng Zhong
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Province Engineering Technology Research Center of Tumor Diagnostic Biomarkers and RNA Interference Drugs, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
46
|
Montemurro M, Monier B, Suzanne M. The mechanical state of pre-tumoral epithelia controls subsequent Drosophila tumor aggressiveness. Dev Cell 2025; 60:1036-1052.e7. [PMID: 39765232 DOI: 10.1016/j.devcel.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/13/2024] [Accepted: 12/04/2024] [Indexed: 04/10/2025]
Abstract
Tumors evolve through the acquisition of increasingly aggressive traits associated with dysplasia. This progression is accompanied by alterations in tumor mechanical properties, especially through extracellular matrix remodeling. However, the contribution of pre-tumoral tissue mechanics to tumor aggressiveness remains poorly known in vivo. Here, we show that adherens junction tension in pre-tumoral tissues dictates subsequent tumor evolution in Drosophila. Increased cell contractility, observed in aggressive tumors before any sign of tissue overgrowth, proved sufficient to trigger dysplasia in normally hyperplastic tumors. In addition, high contractility precedes any changes in cell polarity and contributes to tumor evolution through cell death induction, which favors cell-cell junction weakening. Overall, our results highlight the need to re-evaluate the roles of tumoral cell death and identify pre-tumoral cell mechanics as an unsuspected early marker and key trigger of tumor aggressiveness.
Collapse
Affiliation(s)
- Marianne Montemurro
- Molecular Cellular and Developmental Biology (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31000 Toulouse, France
| | - Bruno Monier
- Molecular Cellular and Developmental Biology (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31000 Toulouse, France.
| | - Magali Suzanne
- Molecular Cellular and Developmental Biology (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31000 Toulouse, France.
| |
Collapse
|
47
|
Zuo S, Li Y, Chen Y, Jiang G, Zhou Z, Ren TB, Chen L, Liu S, Huang S, Zhang XB, Yuan L. Rapid sorting and auxiliary evaluation of malignant breast tumors by accurate imaging analysis of metastasis-related biomarker. SCIENCE ADVANCES 2025; 11:eadr5541. [PMID: 40173246 PMCID: PMC11963997 DOI: 10.1126/sciadv.adr5541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 02/27/2025] [Indexed: 04/04/2025]
Abstract
Accurate differentiation of benign and malignant breast tumors is paramount for establishing schemes of breast cancer treatment and prognosis. Here we report a near-infrared (NIR) fluorescence probe (YF-1) with the overexpressed cathepsin C (CTSC) in metastatic breast tumors as the detecting substrate. This probe allows accurate identification of malignant tumor tissue specimens among tumor tissue specimens with unknown properties in a blind study. Importantly, a series of visible to NIR CTSC-activated fluorescence probes based on the same strategy realize effective identification of malignant tumor tissues, suggesting that CTSC could be the specific identification substrate of malignant breast tumors. Furthermore, a hydrophilic PEG moiety is coupled into YF-1, producing another CTSC-activated NIR probe (YF-2). YF-2 has excellent tumor-targeting capability, enabling the visualization of lung-metastatic breast tumors. The excellent detection accuracy and construction versatility of CTSC probes pave the way for preoperative diagnosis of malignant breast tumors.
Collapse
Affiliation(s)
- Shan Zuo
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Yanhua Li
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Yushi Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Gangwei Jiang
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Zhixuan Zhou
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Tian-Bing Ren
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Lanlan Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Sulai Liu
- Department of Hepatobiliary Surgery/Central Laboratory, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China
| | - Shulin Huang
- Department of Breast and Thyroid Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China
| | - Xiao-Bing Zhang
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Lin Yuan
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| |
Collapse
|
48
|
Bai Z, Zhang G. The application of mendelian randomization in cancer research: a bibliometric analysis. Discov Oncol 2025; 16:463. [PMID: 40185974 PMCID: PMC11971120 DOI: 10.1007/s12672-025-02226-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
Cancer is a major public health and economic issue faced globally today, significantly affecting human health and life. Due to various constraints, exploring the causal relationship between risk factors and cancer is challenging and not exactly accurate. The advent of Mendelian randomization (MR) effectively addresses these issues, providing new avenues for exploring causal relationships. We downloaded literature related to the application of MR in cancer from the Web of Science Core Collection (WoSCC) from 2005 to October 21, 2024, limiting the document type to articles and the language to English, resulting in a total of 2058 articles. We downloaded them in plain text format and extracted information on countries, authors, institutions, keywords, journals, citation counts, and publication dates, utilizing VOSviewer, CiteSpace, and R language for bibliometric analysis. From 2005 to 2024, the number of publications on the application of MR in cancer has shown a growth trend. China was the most productive country (1305); the University of Bristol was the most prolific institution (213); Smith, George Davey published the most articles in this field (59) with a total citation count of 5344; the most prolific journal in this field is Scientific Reports (71). Chronic diseases and cancer, inflammation and cancer, and sex hormones and cancer are three hot topics in the current research on the application of MR in cancer. In the future, optimizing statistical methods, standardizing research processes, collecting data from a broader range of populations, expanding data scale, and integrating other research methods to enhance research quality will be the development trends of MR in cancer research. In summary, this study employed bibliometric methods to comprehensively analyze the literature on the application of MR in cancer over the past 20 years, evaluating the historical development, current applications, research hotspots, and future trends of MR in the field of cancer.
Collapse
Affiliation(s)
- Zhongtao Bai
- Department of General Surgery, Suzhou Hospital of Anhui Medical University, Suzhou, 234000, Anhui Province, China
| | - Genlong Zhang
- Department of General Surgery, Suzhou Hospital of Anhui Medical University, Suzhou, 234000, Anhui Province, China.
| |
Collapse
|
49
|
Bhattacharya R, Avdieiev SS, Bukkuri A, Whelan CJ, Gatenby RA, Tsai KY, Brown JS. The Hallmarks of Cancer as Eco-Evolutionary Processes. Cancer Discov 2025; 15:685-701. [PMID: 40170539 DOI: 10.1158/2159-8290.cd-24-0861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/19/2024] [Accepted: 01/28/2025] [Indexed: 04/03/2025]
Abstract
SIGNIFICANCE Viewing the hallmarks as a sequence of adaptations captures the "why" behind the "how" of the molecular changes driving cancer. This eco-evolutionary view distils the complexity of cancer progression into logical steps, providing a framework for understanding all existing and emerging hallmarks of cancer and developing therapeutic interventions.
Collapse
Affiliation(s)
- Ranjini Bhattacharya
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Cancer Biology, University of South Florida, Tampa, Florida
| | - Stanislav S Avdieiev
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Anuraag Bukkuri
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Center for Evolutionary Biology and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christopher J Whelan
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Robert A Gatenby
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kenneth Y Tsai
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Tumor Microenvironment & Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Joel S Brown
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
50
|
El Ahanidi H, El Azzouzi M, Addoum B, Tetou M, Hassan I, Al Bouzidi A, Oukabli M, Hafidi Alaoui C, Chaoui I, Benbacer L, El Mzibri M, Ameur A, Jandus C, Attaleb M. STAT1 and STAT4 expression as prognostic biomarkers in patients with bladder cancer. Mol Clin Oncol 2025; 22:33. [PMID: 39989605 PMCID: PMC11843079 DOI: 10.3892/mco.2025.2828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/11/2024] [Indexed: 02/25/2025] Open
Abstract
Signal transducer and activator of transcription (STAT) proteins are cytoplasmic transcription factors known to play key roles in numerous physiological and pathological processes, from pathogen response to cancer modulation. However, the roles of some STAT family members, particularly STAT1 and STAT4, in the initiation and progression of bladder cancer (BC) have not been comprehensively studied. The present study investigated the expression pattern of STAT1 and STAT4 in the prognosis and survival of BC taking advantage of patients' specimens and cell lines. In our cohort, high mRNA expression of STAT1 was significantly associated with tumor invasiveness, recurrence and progression, and was shown to increase according to tumor stage in BC cell lines. However, it did not affect patient survival. By contrast, STAT4 exhibited its highest expression in early-stage tumors, without a significant link to the tumor stage. Moreover, it was found that increased STAT4 mRNA expression was associated with improved disease-free survival and overall survival in our cohort. Collectively, these findings suggest that STAT1 and STAT4 could be promising prognostic markers to enhance BC management.
Collapse
Affiliation(s)
- Hajar El Ahanidi
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211 Geneva 4, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, 1066 Lausanne, Switzerland
- Geneva Center for Inflammation Research, 1211 Geneva 4, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, 1211 Geneva 4, Switzerland
- Translational Research Centre in Onco-Hematology (CRTOH), 1211 Geneva 4, Switzerland
| | - Meryem El Azzouzi
- Biology and Medical Research Unit, National Center for Energy, Nuclear Sciences and Techniques, 10010 Rabat, Morocco
| | - Boutaina Addoum
- Biology and Medical Research Unit, National Center for Energy, Nuclear Sciences and Techniques, 10010 Rabat, Morocco
| | - Mohammed Tetou
- Urology Department, Military Hospital of Rabat, 10000 Rabat, Morocco
| | - Ilyass Hassan
- Urology Department, Military Hospital of Rabat, 10000 Rabat, Morocco
| | | | - Mohammed Oukabli
- Urology Department, Military Hospital of Rabat, 10000 Rabat, Morocco
| | - Chaimae Hafidi Alaoui
- Biology and Medical Research Unit, National Center for Energy, Nuclear Sciences and Techniques, 10010 Rabat, Morocco
| | - Imane Chaoui
- Biology and Medical Research Unit, National Center for Energy, Nuclear Sciences and Techniques, 10010 Rabat, Morocco
| | - Laila Benbacer
- Biology and Medical Research Unit, National Center for Energy, Nuclear Sciences and Techniques, 10010 Rabat, Morocco
| | - Mohammed El Mzibri
- Biology and Medical Research Unit, National Center for Energy, Nuclear Sciences and Techniques, 10010 Rabat, Morocco
| | - Ahmed Ameur
- Urology Department, Military Hospital of Rabat, 10000 Rabat, Morocco
| | - Camilla Jandus
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211 Geneva 4, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, 1066 Lausanne, Switzerland
- Geneva Center for Inflammation Research, 1211 Geneva 4, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), University of Geneva, 1211 Geneva 4, Switzerland
- Translational Research Centre in Onco-Hematology (CRTOH), 1211 Geneva 4, Switzerland
| | - Mohammed Attaleb
- Biology and Medical Research Unit, National Center for Energy, Nuclear Sciences and Techniques, 10010 Rabat, Morocco
| |
Collapse
|