1
|
Jiang J, Peng W, Sun N, Zhao D, Cui W, Lai Y, Zhang C, Duan C, Zeng W. Unraveling the anoikis-cancer nexus: a bibliometric analysis of research trends and mechanisms. Future Sci OA 2025; 11:2484159. [PMID: 40160087 PMCID: PMC11959893 DOI: 10.1080/20565623.2025.2484159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Cancer, influenced by genetics and the environment, involves anoikis, a cell death mechanism upon extracellular matrix detachment crucial for metastasis. Understanding this relationship is key for therapy. We analyze cancer and anoikis trends using bibliometrics. METHODS A search was conducted from Web of Science Core, PubMed, Scopus and non-English databases such as the CNKI (inception- 21 December 2024). Data analysis employed Microsoft Excel, VOSviewer, CiteSpace, R software, and the online platform (https://bibliometric.com/). RESULTS 2510 publications were retrieved, with a significant increase in the last decade. China led, the University of Texas system was productive, and the Oncogene Journal was popular. Breast, and colorectal cancers were frequently studied. Among them, representative tumor-related mechanisms were identified, commonalities such as (EMT, ECM, autophagy) and respective specific mechanisms were summarized. CONCLUSION This bibliometric analysis highlights rapid advances in anoikis research in cancer, emphasizing EMT and FAK pathways' translational potential, guiding targeted therapies, and improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Junjie Jiang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Wei Peng
- Department of Oncology, Hunan Provincial People’s Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, People’s Republic of China
| | - Nianzhe Sun
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Deze Zhao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Weifang Cui
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Yuwei Lai
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
| | - Chaojun Duan
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
- Institute of Medical Sciences, Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Wei Zeng
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagnosis & Treatment, Changsha, Hunan, People’s Republic of China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
2
|
Zheng L, Li K, Tang X, Li C, Nie H, Han L, Li Y. A microfluidic co-culture platform for lung cancer cells electrotaxis study under the existence of stromal cells. Bioelectrochemistry 2025; 164:108917. [PMID: 39904303 DOI: 10.1016/j.bioelechem.2025.108917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/24/2025] [Accepted: 01/25/2025] [Indexed: 02/06/2025]
Abstract
Tumor metastasis is an important reason for the poor prognosis and high mortality in cancer patients. As major component of stromal cells in tumor microenvironment, cancer-associated fibroblasts (CAFs) secreted various factors to promote tumor metastasis. Studies have indicated that endogenous direct current electric field (dcEF) around tumor tissue induced directional migration of cancer cells. However, the regulatory effect of CAFs on cancer migration under dcEF stimulation is still unknown. In this study, a two-layers polydimethylsiloxane (PDMS)-based microfluidic chip was fabricated. The introduction of concave structures achieved the non-contacted co-culture of different cell types, and parallel channels in the chip provided stable and homogeneous dcEF. Cells electrotactic response was evaluated under co-culture circumstance. The results showed that CAFs exhibited directional migration towards anode under dcEF stimulation, while A549 cells had a trend of directional migration towards cathode. The co-existence of CAFs and dcEF significantly enhanced the motility and cathodal migration of A549 cells, suggesting synergistic influences of chemotaxis from CAFs and electrotaxis from dcEF stimulation. Moreover, we demonstrated that lung normal fibroblasts acquired CAFs properties after stimulated by dcEF, characterizing by increasing gene expression of α-SMA and IL-6. Overall, Our device and study provide new insight for tumor electrotaxis in complex microenvironment.
Collapse
Affiliation(s)
- Lina Zheng
- Hebei Key Laboratory of Public Health Safety, School of Public Health, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding 071002, PR China
| | - Keying Li
- Hebei Key Laboratory of Public Health Safety, School of Public Health, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding 071002, PR China
| | - Xianmei Tang
- Hebei Key Laboratory of Public Health Safety, School of Public Health, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding 071002, PR China
| | - Cuiping Li
- Hebei Key Laboratory of Public Health Safety, School of Public Health, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding 071002, PR China
| | - Hailiang Nie
- Hebei Key Laboratory of Public Health Safety, School of Public Health, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding 071002, PR China
| | - Lirong Han
- Hebei Key Laboratory of Public Health Safety, School of Public Health, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding 071002, PR China.
| | - Yaping Li
- Hebei Key Laboratory of Public Health Safety, School of Public Health, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding 071002, PR China.
| |
Collapse
|
3
|
Rajalekshmi R, Agrawal DK. Transcriptional and post-translational mechanisms of ECM remodeling in rotator cuff tendons under hyperlipidemic conditions. Life Sci 2025; 372:123647. [PMID: 40246193 DOI: 10.1016/j.lfs.2025.123647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/11/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Rotator cuff injuries present significant clinical challenges, often resulting in chronic pain and functional impairment. In this study, we examined the effects of hyperlipidemia (HYP), a systemic metabolic condition, on tendon health. Histological analysis of infraspinatus tendons from hyperlipidemic swine revealed well-organized extracellular matrix (ECM) structures, comparable to those in non-hyperlipidemic (NONHYP) animals, suggesting ECM reorganization. Upstream SIGNOR3.0 analysis demonstrated that tumor necrosis factor receptor-associated factor 6 (TRAF6) activates transcription factor Yin Yang 1 (YY1) via kinase signaling, underscoring its role in tendon ECM remodeling. Hence, we futher examined the role of YY1, which is a critical regulator of collagen synthesis identified through network analysis. Although TRAF6 levels remained unchanged in HYP conditions, increased YY1 expression correlated with elevated COL1 gene expression. Additionally, twist-related protein 1 (TWIST1) emerged as another key molecule, existing in both homo- and heterodimer forms in NON-HYP conditions, but only as a heterodimer in HYP. YY1 enhanced COL1 transcription in the hyperlipidemic environment, while TWIST1 heterodimer formation facilitated collagen crosslinking. Notably, increased YY1 expression inhibited MMP3, resulting in the inactivity of MMP1, MMP8, and MMP9, thereby preserving collagen levels. These findings highlight the complex molecular interactions involving transcriptional regulation by YY1 and post-translational regulation by the TWIST1 heterodimer, essential for the deposition of mature collagen fibrils and driving tendon remodeling in hyperlipidemic conditions. This study offers valuable insights for the change of tendon health condition in hyperlipidemia disease or tendon pathology.
Collapse
Affiliation(s)
- Resmi Rajalekshmi
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA.
| |
Collapse
|
4
|
Shahraki FH, Shareghi B, Farhadian S. Deciphering the molecular interaction between Vitamin D3 and pepsin by in vitro and in silico perspectives. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 334:125956. [PMID: 40024088 DOI: 10.1016/j.saa.2025.125956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
The current study explored the molecular interaction between Vitamin D3 (Vit D3) and pepsin using multi-spectroscopic, molecular dynamic simulation (MDS), and molecular docking. The fluorescence emission spectra discovered Vit D3 interacted with pepsin in a static quenching manner due to the formation of the steady-state complex. Thermodynamic data revealed the spontaneous binding of Vit D3 on pepsin. The formation of the Pepsin-Vit D3 complex was also validated by circular dichroism (CD) spectroscopy. The fluorescence and CD spectroscopy results revealed Vit D3 altered the tertiary and secondary structure of pepsin, respectively. Meanwhile, FTIR spectroscopy results revealed a hypochromic shift in the amide I and II peaks. Kinetic parameters showed Vit D3 inhibited the activity of pepsin by the uncompetitive process. Applied spectroscopic methods disclosed that Vit D3 binding to pepsin caused microenvironmental modifications around the aromatic residues of protein and changed its structure and function. Moreover, MD simulation and molecular docking were done to analyze the formation of Pepsin-Vit D3 complexes. Molecular docking findings demonstrated the interaction of Vit D3 with pepsin mainly involved van der Waals forces and hydrogen bonds that were in good agreement with the fluorescence results. Finally, MDS findings including RMSD, RMSF, and RG confirmed all the experimental data.
Collapse
Affiliation(s)
- Fatemeh Hashemi Shahraki
- Department of Biology, Faculty of Science, Shahrekord University, Shahrekord, P.O. Box.115, Iran; Central Laboratory, Shahrekord University, Shahrekord, Iran
| | - Behzad Shareghi
- Department of Biology, Faculty of Science, Shahrekord University, Shahrekord, P.O. Box.115, Iran; Central Laboratory, Shahrekord University, Shahrekord, Iran.
| | - Sadegh Farhadian
- Department of Biology, Faculty of Science, Shahrekord University, Shahrekord, P.O. Box.115, Iran; Central Laboratory, Shahrekord University, Shahrekord, Iran.
| |
Collapse
|
5
|
Al Azim M, Di Martino JS. ECM, integrins, and DDRs: A nexus of cancer progression, therapy, and future directions. Matrix Biol 2025; 138:27-43. [PMID: 40350240 DOI: 10.1016/j.matbio.2025.04.002] [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: 01/29/2025] [Revised: 03/14/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025]
Abstract
Collagen is the most abundant protein in mammals, significantly contributing to cancer progression. Cells express two primary well-conserved collagen receptors, integrins and discoidin domain receptors (DDRs), which bind collagen on distinct sites, suggesting that cancer cells must integrate both signals to decide their fate. The crosstalk between integrins and DDRs mediated by collagen binding produces dynamic, integrated signals that control tumor progression, therapeutic resistance, and cancer cell heterogeneity. This review will discuss the dynamic interplay among collagen, integrins, and DDRs in ECM remodeling during cancer progression and these receptors' crosstalk. In addition, we explored current and future directions for ECM receptor-targeted therapies, including nanotechnologies and precision medicine, to improve therapeutic outcomes by establishing a proper balance between integrins and DDRs in cancer.
Collapse
Affiliation(s)
- Md Al Azim
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla 10595, NY, USA
| | - Julie S Di Martino
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla 10595, NY, USA.
| |
Collapse
|
6
|
Cui Y, Cui Y, Ding Y, Nakai K, Wei L, Le Y, Ye X, Sakurai T. OmniClust: A versatile clustering toolkit for single-cell and spatial transcriptomics data. Methods 2025; 238:84-94. [PMID: 40057293 DOI: 10.1016/j.ymeth.2025.03.007] [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: 01/31/2025] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
In recent years, RNA transcriptome sequencing technology has been continuously evolving, ranging from single-cell transcriptomics to spatial transcriptomics. Although these technologies are all based on RNA sequencing, each sequencing technology has its own unique characteristics, and there is an urgent need to develop an algorithmic toolkit that integrates both sequencing techniques. To address this, we have developed OmniClust, a toolkit based on single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics data. OmniClust employs deep learning algorithms for feature learning and clustering of spatial transcriptomics data, while utilizing machine learning algorithms for clustering scRNA-seq data. OmniClust was tested on 12 spatial transcriptomics benchmark datasets, demonstrating high clustering accuracy across multiple clustering evaluation metrics. It was also evaluated on four scRNA-seq benchmark datasets, achieving high clustering accuracy based on various clustering evaluation metrics. Furthermore, we applied OmniClust to downstream analyses of spatial transcriptomics and single-cell RNA breast cancer data, showcasing its potential to uncover and interpret the biological significance of cancer transcriptome data. In summary, OmniClust is a clustering tool designed for both single-cell transcriptomics and spatial transcriptomics data, demonstrating outstanding performance.
Collapse
Affiliation(s)
- Yaxuan Cui
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan
| | - Yang Cui
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Yi Ding
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Kenta Nakai
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan; Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Leyi Wei
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao SAR, China
| | - Yuyin Le
- Department of Radiation Oncology Fuzhou Pulmonary Hospital of Fujian Province , Teaching Hospital of Fujian Medical University, China.
| | - Xiucai Ye
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan.
| | - Tetsuya Sakurai
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan
| |
Collapse
|
7
|
Jomova K, Alomar SY, Valko R, Liska J, Nepovimova E, Kuca K, Valko M. Flavonoids and their role in oxidative stress, inflammation, and human diseases. Chem Biol Interact 2025; 413:111489. [PMID: 40147618 DOI: 10.1016/j.cbi.2025.111489] [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: 06/02/2024] [Revised: 02/23/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Oxidative stress and chronic inflammation are important drivers in the pathogenesis and progression of many chronic diseases, such as cancers of the breast, kidney, lung, and others, autoimmune diseases (rheumatoid arthritis), cardiovascular diseases (hypertension, atherosclerosis, arrhythmia), neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, Huntington's disease), mental disorders (depression, schizophrenia, bipolar disorder), gastrointestinal disorders (inflammatory bowel disease, colorectal cancer), and other disorders. With the increasing demand for less toxic and more tolerable therapies, flavonoids have the potential to effectively modulate the responsiveness to conventional therapy and radiotherapy. Flavonoids are polyphenolic compounds found in fruits, vegetables, grains, and plant-derived beverages. Six of the twelve structurally different flavonoid subgroups are of dietary significance and include anthocyanidins (e.g. pelargonidin, cyanidin), flavan-3-ols (e.g. epicatechin, epigallocatechin), flavonols (e.g. quercetin, kaempferol), flavones (e.g. luteolin, baicalein), flavanones (e.g. hesperetin, naringenin), and isoflavones (daidzein, genistein). The health benefits of flavonoids are related to their structural characteristics, such as the number and position of hydroxyl groups and the presence of C2C3 double bonds, which predetermine their ability to chelate metal ions, terminate ROS (e.g. hydroxyl radicals formed by the Fenton reaction), and interact with biological targets to trigger a biological response. Based on these structural characteristics, flavonoids can exert both antioxidant or prooxidant properties, modulate the activity of ROS-scavenging enzymes and the expression and activation of proinflammatory cytokines (e.g., interleukin-1beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α)), induce apoptosis and autophagy, and target key signaling pathways, such as the nuclear factor erythroid 2-related factor 2 (Nrf2) and Bcl-2 family of proteins. This review aims to briefly discuss the mutually interconnected aspects of oxidative and inflammatory mechanisms, such as lipid peroxidation, protein oxidation, DNA damage, and the mechanism and resolution of inflammation. The major part of this article discusses the role of flavonoids in alleviating oxidative stress and inflammation, two common components of many human diseases. The results of epidemiological studies on flavonoids are also presented.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nitra, 949 74, Slovakia
| | - Suliman Y Alomar
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Richard Valko
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Jan Liska
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, 811 08, Bratislava, Slovakia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic; Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, Ostrava-Poruba, 708 00, Czech Republic
| | - Kamil Kuca
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, Ostrava-Poruba, 708 00, Czech Republic; Biomedical Research Center, University Hospital Hradec Kralove, 5005, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia.
| |
Collapse
|
8
|
Xu S, Zhang H, Tian Y. Pericytes in hematogenous metastasis: mechanistic insights and therapeutic approaches. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01073-6. [PMID: 40392500 DOI: 10.1007/s13402-025-01073-6] [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: 02/20/2025] [Accepted: 05/09/2025] [Indexed: 05/22/2025] Open
Abstract
Metastasis, the leading cause of cancer-related deaths, underscores the critical need to understand its regulatory mechanisms to improve prevention and treatment strategies for late-stage tumors. Hematogenous dissemination is a key route of metastasis. However, as the gatekeeper of vessels, the role of pericytes in hematogenous metastasis remains largely unknown. In this review, we comprehensively explore the contributions of pericytes throughout the metastatic cascade, particularly their functions that extend beyond influencing tumor angiogenesis. Pericytes should not be perceived as passive bystanders, but rather as active participants in various stages of the metastatic cascade. Pericytes-targeted therapy may provide novel insights for preventing and treating advanced-stage tumor.
Collapse
Affiliation(s)
- Shuo Xu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| | - Hong Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China.
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China.
| |
Collapse
|
9
|
Kjær A, Kristjánsdóttir N, Juul RI, Nordentoft I, Birkenkamp-Demtröder K, Ahrenfeldt J, Strandgaard T, Radif D, Hodgson D, Abbosh C, Aerts HJWL, Agerbæk M, Jensen JB, Birkbak NJ, Dyrskjøt L. Low T cell diversity associates with poor outcome in bladder cancer: A comprehensive longitudinal analysis of the T cell receptor repertoire. Cell Rep Med 2025; 6:102101. [PMID: 40315845 DOI: 10.1016/j.xcrm.2025.102101] [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: 08/30/2024] [Revised: 11/20/2024] [Accepted: 04/09/2025] [Indexed: 05/04/2025]
Abstract
T cells are crucial effector cells in the endogenous defense against cancer, yet the clinical impact of their quantity, diversity, and dynamics remains underexplored. Here, we investigate the clinical relevance of the T cell receptor (TCR) repertoire in patients with bladder cancer. In advanced-stage disease, low pre-treatment peripheral TCR diversity is associated with worse overall survival (p = 0.024), particularly when coupled with low circulating T cell fractions (p = 0.00049). These low-diversity repertoires are dominated by hyper-expanded clones that persist throughout treatment. Further longitudinal analysis reveals reductions in TCR diversity after treatment, indicating adverse effects on the immune system. In early-stage disease, immunotherapy increases TCR diversity in patients with good outcomes. Furthermore, single-cell sequencing identifies most hyper-expanded clones as cytotoxic T cells, while non-expanded clones are predominantly naive T cells. Overall, this highlights TCR diversity as a promising biomarker, offering opportunities for tailored oncological treatments to enhance clinical outcomes.
Collapse
Affiliation(s)
- Asbjørn Kjær
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Nanna Kristjánsdóttir
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Randi Istrup Juul
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Iver Nordentoft
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark
| | - Karin Birkenkamp-Demtröder
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Johanne Ahrenfeldt
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark
| | - Trine Strandgaard
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Deema Radif
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Darren Hodgson
- Cancer Biomarker Development, Oncology R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Christopher Abbosh
- Cancer Biomarker Development, Oncology R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Hugo J W L Aerts
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA 02114, USA; Radiology and Nuclear Medicine, CARIM & GROW, Maastricht University, 6200 MD Maastricht, the Netherlands
| | - Mads Agerbæk
- Department of Oncology, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark
| | - Jørgen Bjerggaard Jensen
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark; Department of Urology, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark
| | - Nicolai J Birkbak
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark.
| | - Lars Dyrskjøt
- Department of Molecular Medicine, Aarhus University Hospital, 8200 Aarhus N, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark.
| |
Collapse
|
10
|
Pedersen RS, Hettich A, Thorlacius-Ussing J, Langholm LL, Crespo-Bravo M, Chen IM, Hansen CP, Johansen JS, Diab HMH, Jorgensen LN, Karsdal M, Willumsen N. Proteolytic degradation of Beta-Ig H3 (βigH3/TGFBI) can be quantified non-invasively in serum and predicts prognosis in patients with advanced pancreatic ductal adenocarcinoma. BMC Cancer 2025; 25:905. [PMID: 40394523 DOI: 10.1186/s12885-025-14283-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025] Open
Abstract
The extracellular matrix (ECM) protein Beta-Ig H3 (βigH3, also known as transforming growth factor β induced protein (TGFBI)) is related to poor prognosis in patients with pancreatic ductal adenocarcinoma (PDAC). Proteolytic cleavage of βigH3 has been shown to result in release of the N-terminal fragment covering amino acid 1 to 137, but whether the degradation of βigH3 is associated to prognosis has yet to be determined. In this study we developed an ELISA targeting a collagenase generated fragment of βigH3 (cβigH3) in human serum to use the fragment as a biomarker reflecting degradation of βigH3. We demonstrated that the assay was specific to the cleaved fragment (cβigH3) and confirmed the generation of cβigH3 from degradation of fibroblast generated matrices. Moreover, higher levels of cβigH3 were released upon degradation of matrices produced by TGF-β stimulated pancreatic fibroblast compared to matrices produced by pancreatic fibroblast without TGF-β stimulation, indicating an association of the biomarker with degradation of fibrotic matrix. To evaluate the clinical relevance, we first measured cβigH3 in a cohort of 220 patients with different types of cancer with detectable levels for all 11 cancer types. We then measured the cβigH3 biomarker in pre-treatment serum from a cohort of 469 patients with locally advanced or metastatic PDAC and found that high levels of cβigH3 were associated with longer overall survival independently of age, disease stage, performance status, carbohydrate antigen 19-9 (CA19-9), and the tumor fibrosis biomarker PRO-C3 as compared to patients with high levels of cβigH3 (HR 0.78, 95% CI: 0.0.61-0.98, p = 0.04). In conclusion, cβigH3 reflects proteolytic degradation of βigH3 and shows potential as an independent prognostic biomarker for patients with advanced PDAC.
Collapse
Affiliation(s)
- Rasmus S Pedersen
- Nordic Bioscience A/S, 2730, Herlev, Denmark.
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| | - Annika Hettich
- Nordic Bioscience A/S, 2730, Herlev, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | | | | | - Marina Crespo-Bravo
- Nordic Bioscience A/S, 2730, Herlev, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Inna M Chen
- Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, 2730, Herlev, Denmark
| | - Carsten P Hansen
- Department of Surgery, Copenhagen University Hospital - Rigshospitalet, 2100, Copenhagen, Denmark
| | - Julia S Johansen
- Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, 2730, Herlev, Denmark
- Department of Medicine, Copenhagen University Hospital - Herlev and Gentofte, 2730, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Hadi M H Diab
- Digestive Disease Center, Copenhagen University Hospital - Bispebjerg and Frederiksberg, 2400, Copenhagen, Denmark
| | - Lars N Jorgensen
- Digestive Disease Center, Copenhagen University Hospital - Bispebjerg and Frederiksberg, 2400, Copenhagen, Denmark
| | | | | |
Collapse
|
11
|
Zhou X, Wang J, Lu M, Fang L, Zhao J, Li D. Leucine-rich repeat-containing 56 promotes breast cancer progression via modulation of the RhoA/ROCKs signaling axis. MOLECULAR BIOMEDICINE 2025; 6:31. [PMID: 40388100 PMCID: PMC12089637 DOI: 10.1186/s43556-025-00271-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 04/18/2025] [Accepted: 04/28/2025] [Indexed: 05/20/2025] Open
Abstract
Breast cancer is one of the most common malignancies with a poor five-year survival rate with metastatic disease among women. It has well been documented that leucine-rich repeat-containing (LRRC) family of proteins are remarkably and aberrantly dysregulated across diverse cancer types. Notably, leucine-rich repeat-containing 56 (LRRC56) was found upregulated in metastatic breast cancer, and plays a crucial role for the movement of cilia via intraflagellar transport 88 (IFT88). However, the role for LRRC56 in breast cancer progression and regulation of IFT88 and associated pathways in metastatic progression of breast cancer has not been defined. Via in vitro functional assessments, we found that LRRC56 pivotally influences the proliferative, migratory and invasive capabilities of cancer cells. Further, via in-vivo assessments, we demonstrated that downregulation of LRRC56 effectively inhibits the growth of breast cancer xenograft tumors and their metastasis to the lungs. Mechanistically, we found that LRRC56 interacts with IFT88 to regulate yes-associated protein 1 (YAP1) expression via modulating the Ras homolog family member A (RhoA)/ Rho-associated protein kinases (ROCKs) signaling pathway. LRRC56 also regulates the expression of integrins and several other key molecules including MMP2, MMP9, FAK, as well as markers of epithelial-mesenchymal transition such as E-cadherin and N-cadherin. In summary, our results demonstrate that overexpression of LRRC56 promotes breast cancer progression via upregulating IFT88/YAP1-RhoA/ROCKs pathway, reprogramming extracellular matrix, and enhancing epithelial-mesenchymal transition. These findings highlight a critical role of LRRC56 in promoting breast cancer progression, suggesting that targeting of LRRC56 may offer a promising strategy for treating metastatic breast cancer.
Collapse
Affiliation(s)
- Xiqian Zhou
- Department of Breast and Thyroid Surgery,Institute of Breast Disease, Shanghai Tenth People's Hospital, Tongji University School of Medicine, NO.301 Yanchang Middle Road, Shanghai, 200072, People's Republic of China
| | - Jiaxin Wang
- Department of Breast and Thyroid Surgery,Institute of Breast Disease, Shanghai Tenth People's Hospital, Tongji University School of Medicine, NO.301 Yanchang Middle Road, Shanghai, 200072, People's Republic of China
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Meiling Lu
- Department of Central Laboratory, School of Life Sciences and Technology, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200072, China
| | - Lin Fang
- Department of Breast and Thyroid Surgery,Institute of Breast Disease, Shanghai Tenth People's Hospital, Tongji University School of Medicine, NO.301 Yanchang Middle Road, Shanghai, 200072, People's Republic of China
| | - Junyong Zhao
- Department of Breast and Thyroid Surgery,Institute of Breast Disease, Shanghai Tenth People's Hospital, Tongji University School of Medicine, NO.301 Yanchang Middle Road, Shanghai, 200072, People's Republic of China.
| | - Dengfeng Li
- Department of Breast and Thyroid Surgery,Institute of Breast Disease, Shanghai Tenth People's Hospital, Tongji University School of Medicine, NO.301 Yanchang Middle Road, Shanghai, 200072, People's Republic of China.
| |
Collapse
|
12
|
Li Y, Zhang R, Dang Y, Liang Y, Wang L, Chen N, Zhuang L, Liu W, Gong T. Sieging tumor cells using an amorphous ferric coordination polymer. MATERIALS HORIZONS 2025; 12:3388-3398. [PMID: 40025991 DOI: 10.1039/d4mh01558d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Metastasis is one of the main reasons for cancer treatment failure. Unfortunately, most treatment approaches inevitably damage the extracellular matrix (ECM) during tumor cell elimination, thereby augmenting the risk of metastasis. Herein, we proposed a "sieging tumor cells" strategy based on ferric coordination polymers (FeCPs), which involved anchoring tumor cells through ECM consolidation and selectively eliminating them in the tumor regions. Due to the weak coordination interactions and amorphous structure of FeCPs, the acidic tumor microenvironment facilitated their disintegration, releasing salicylic acid (SA), 2,5-dihydroxyterephthalic acid (DHTA) and Fe3+ ions. The released SA inhibited heparinase activity to consolidate the ECM, while Fe-mediated chemodynamic therapy (CDT) was enhanced by DHTA due to its fast electron transport behavior, ultimately inhibiting tumor growth and metastasis. The results from the orthotopic 4T1 breast tumor model indicated that lung metastasis was reduced by about 90%, and the survival rate improved by 70% after FeCP treatment. Overall, this "sieging tumor cells" strategy provides an emerging approach for the treatment of malignant tumors by consolidating the ECM in combination with self-enhanced CDT.
Collapse
Affiliation(s)
- Yanli Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Ruoqi Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Yuanye Dang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Yongyu Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Lulu Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Na Chen
- Soochow University Library, Soochow University, Suzhou 215006, China
| | - Luwen Zhuang
- Center for Water Resources and Environment, and Guangdong Key Laboratory of Marine Civil Engineering, School of Civil Engineering, Sun Yat-sen University, Guangzhou 510275, China.
| | - Wen Liu
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, China.
| | - Teng Gong
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
13
|
Liu L, Wang H, Chen R, Song Y, Wei W, Baek D, Gillin M, Kurabayashi K, Chen W. Cancer-on-a-chip for precision cancer medicine. LAB ON A CHIP 2025. [PMID: 40376718 DOI: 10.1039/d4lc01043d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Many cancer therapies fail in clinical trials despite showing potent efficacy in preclinical studies. One of the key reasons is the adopted preclinical models cannot recapitulate the complex tumor microenvironment (TME) and reflect the heterogeneity and patient specificity in human cancer. Cancer-on-a-chip (CoC) microphysiological systems can closely mimic the complex anatomical features and microenvironment interactions in an actual tumor, enabling more accurate disease modeling and therapy testing. This review article concisely summarizes and highlights the state-of-the-art progresses in CoC development for modeling critical TME compartments including the tumor vasculature, stromal and immune niche, as well as its applications in therapying screening. Current dilemma in cancer therapy development demonstrates that future preclinical models should reflect patient specific pathophysiology and heterogeneity with high accuracy and enable high-throughput screening for anticancer drug discovery and development. Therefore, CoC should be evolved as well. We explore future directions and discuss the pathway to develop the next generation of CoC models for precision cancer medicine, such as patient-derived chip, organoids-on-a-chip, and multi-organs-on-a-chip with high fidelity. We also discuss how the integration of sensors and microenvironmental control modules can provide a more comprehensive investigation of disease mechanisms and therapies. Next, we outline the roadmap of future standardization and translation of CoC technology toward real-world applications in pharmaceutical development and clinical settings for precision cancer medicine and the practical challenges and ethical concerns. Finally, we overview how applying advanced artificial intelligence tools and computational models could exploit CoC-derived data and augment the analytical ability of CoC.
Collapse
Affiliation(s)
- Lunan Liu
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
| | - Huishu Wang
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
| | - Ruiqi Chen
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - Yujing Song
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
| | - William Wei
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - David Baek
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - Mahan Gillin
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - Katsuo Kurabayashi
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
- Department of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA.
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
- Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
14
|
Baldassarre G, L de la Serna I, Vallette FM. Death-ision: the link between cellular resilience and cancer resistance to treatments. Mol Cancer 2025; 24:144. [PMID: 40375296 PMCID: PMC12080166 DOI: 10.1186/s12943-025-02339-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/22/2025] [Indexed: 05/18/2025] Open
Abstract
One of the key challenges in defeating advanced tumors is the ability of cancer cells to evade the selective pressure imposed by chemotherapy, targeted therapies, immunotherapy and cellular therapies. Both genetic and epigenetic alterations contribute to the development of resistance, allowing cancer cells to survive initially effective treatments. In this narration, we explore how genetic and epigenetic regulatory mechanisms influence the state of tumor cells and their responsiveness to different therapeutic strategies. We further propose that an altered balance between cell growth and cell death is a fundamental driver of drug resistance. Cell death programs exist in various forms, shaped by cell type, triggering factors, and microenvironmental conditions. These processes are governed by temporal and spatial constraints and appear to be more heterogeneous than previously understood. To capture the intricate interplay between death-inducing signals and survival mechanisms, we introduce the concept of Death-ision. This framework highlights the dynamic nature of cell death regulation, determining whether specific cancer cell clones evade or succumb to therapy. Building on this understanding offers promising strategies to counteract resistant clones and enhance therapeutic efficacy. For instance, combining DNMT inhibitors with immune checkpoint blockade may counteract YAP1-driven resistance or the use of transcriptional CDK inhibitors could prevent or overcome chemotherapy resistance. Death-ision aims to provide a deeper understanding of the diversity and evolution of cell death programs, not only at diagnosis but also throughout disease progression and treatment adaptation.
Collapse
Affiliation(s)
- Gustavo Baldassarre
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, 33081, Italy.
| | - Ivana L de la Serna
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA.
| | - François M Vallette
- Centre de Recherche en Cancérologie et Immunologie Intégrées Nantes Angers (CRCI2 NA), INSERM UMR1307/CNRS UMR 6075/Nantes Université/Univ. Angers. Nantes, 44007, Nantes, France.
- Institut de Cancérologie de L'Ouest (ICO), 44085, Saint-Herblain, France.
| |
Collapse
|
15
|
Zhang W, Li JB, Liu HM, Wang KM, Xiao BL, Wang YM, Liang JJ, Zeng J, Zhang LZ, Feng YYF, Fu QY, Wang XX, Liu YT, Cheng XX, Li J, Zhang YY, Zhang G, Zhang JL, Yu ZL, Shao Z, Xiong XP, Jia J, Liu B, Chen G. PERK+ Macrophages Drive Immunotherapy Resistance in Lymph Node Metastases of Oral Squamous Cell Carcinoma. Clin Cancer Res 2025; 31:1894-1911. [PMID: 40036693 DOI: 10.1158/1078-0432.ccr-24-3135] [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: 09/23/2024] [Revised: 12/06/2024] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
PURPOSE Neoadjuvant anti-PD-1 immunotherapy combined with chemotherapy has shown promising pathologic responses in various cancers, including oral squamous cell carcinoma (OSCC). However, the pathologic response of lymph node (LN) metastases remains poorly understood. This study aims to systematically evaluate the pathologic response rates (pRR) of LN metastases in patients with OSCC and identify potential targets to improve therapeutic outcomes. PATIENTS AND METHODS We assessed the pRRs of LN metastases and matched primary tumors (PT) in patients with OSCC enrolled in a randomized, two-arm, phase II clinical trial (NCT04649476). Single-cell and spatial transcriptomics and multiplex IHC were performed to analyze the tumor microenvironment and identify potential therapeutic targets in LN metastases. A neoadjuvant orthotopic OSCC mouse model was established to evaluate the therapeutic potential of these targets. RESULTS We observed significant heterogeneity in pathologic regression of LN metastases, with lower pRRs compared with PTs. pRRs in LN metastases were correlated with overall and disease-free survival in patients with OSCC. We identified an abundance of macrophages in LN metastases exhibiting an unfolded protein response and activated protein kinase RNA-like endoplasmic reticulum kinase (PERK) signaling. These macrophages contributed to an extracellular matrix-enriched microenvironment through interactions with fibroblasts, which hindered T cell-mediated cytotoxicity. Pharmacologic inhibition of the PERK pathway significantly enhanced anti-PD-1 therapy in LN metastases and PTs in the mouse model. CONCLUSIONS Our study confirms that the pathologic response of LN metastases in patients with OSCC undergoing neoadjuvant immunotherapy or immunochemotherapy is inferior to that of PTs. It suggests that targeting the PERK pathway in macrophages could be a potential strategy to enhance treatment outcomes.
Collapse
Affiliation(s)
- Wei Zhang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jin-Bang Li
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hai-Ming Liu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Kui-Ming Wang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bo-Lin Xiao
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi-Man Wang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jia-Jie Liang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jun Zeng
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lin-Zhou Zhang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang-Ying-Fan Feng
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qiu-Yun Fu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xin-Xin Wang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yu-Tong Liu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xiao-Xia Cheng
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jing Li
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yu-Ying Zhang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Gao Zhang
- Faculty of Dentistry, The University of Hong Kong, Sai Ying Pun, Hong Kong
| | - Jia-Li Zhang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral Pathology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zi-Li Yu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhe Shao
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xue-Peng Xiong
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jun Jia
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bing Liu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Gang Chen
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| |
Collapse
|
16
|
Lee J, Cho H, Kim J, Lim J, Kang Y, Kim WJ. Breaking barriers: Nitric oxide-releasing nanocomplexes for collagen degradation and enhanced αPD-L1 immunotherapy in deep tumor. J Control Release 2025; 381:113576. [PMID: 40010409 DOI: 10.1016/j.jconrel.2025.02.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/08/2025] [Accepted: 02/23/2025] [Indexed: 02/28/2025]
Abstract
Overcoming the physical barrier of the extracellular matrix (ECM) surrounding tumors is a critical challenge in achieving effective immune checkpoint blockade (ICB). The dense ECM impedes the infiltration of immune checkpoint inhibitors (ICIs) and cytotoxic T lymphocytes (CTLs) into tumor tissues. To address this, we design a nanocomplex incorporating a reactive oxygen species (ROS)-responsive nitric oxide (NO) prodrug around TANNylated αPD-L1. Within the tumor microenvironment (TME), this nanocomplex accumulates and selectively releases NO in response to ROS. The released NO activates matrix metalloproteinases (MMPs) in the ECM, leading to collagen degradation. Following this, the pH-responsive release of αPD-L1 in the deeper tumor regions ensures effective delivery, allowing CTLs to penetrate the tumor more efficiently by bypassing the ECM barrier, thereby enhancing immunotherapy. Overall, this study applies a nanocomplex capable of releasing NO and αPD-L1 in the tumor to a solid tumor model, successfully inhibiting tumor growth by altering the immunosuppressive environment through improved penetration.
Collapse
Affiliation(s)
- Jihye Lee
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute (POSTECH), Pohang 37673, South Korea
| | - Hyoeun Cho
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute (POSTECH), Pohang 37673, South Korea
| | - Jieun Kim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute (POSTECH), Pohang 37673, South Korea
| | - Junha Lim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute (POSTECH), Pohang 37673, South Korea
| | - Yeoul Kang
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute (POSTECH), Pohang 37673, South Korea
| | - Won Jong Kim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute (POSTECH), Pohang 37673, South Korea.
| |
Collapse
|
17
|
Wang C, Yu X, Teer JK, Yao J, Du D, Liu X, Thompson ZJ, Wang MH, Welsh EA, Memon D, Chan TA, Makarov V, Anadon CM, Saeed L, Boyle TA, Fang B, Koomen JM, Cox C, Landin AM, Yoder SJ, Kim S, Chen DT, Pilon-Thomas SA, Conejo-Garcia JR, Antonia SJ, Haura EB, Creelan BC. Impaired T cell and neoantigen retention in time-serial analysis of metastatic non-small cell lung cancer in patients unresponsive to TIL cell therapy. NATURE CANCER 2025:10.1038/s43018-025-00946-x. [PMID: 40341231 DOI: 10.1038/s43018-025-00946-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/10/2025] [Indexed: 05/10/2025]
Abstract
Cell therapy with tumor-infiltrating lymphocytes (TILs) has yielded durable responses for multiple cancer types, but the causes of therapeutic resistance remain largely unknown. Here multidimensional analysis was performed on time-serial tumor and blood in a lung cancer TIL therapy trial. Using T cell receptor sequencing on both functionally expanded T cells and neoantigen-loaded tetramer-sorted T cells, we identified tumor antigen-specific T cell receptors. We then mapped clones into individual transcriptomes and found that tumor-reactive clonotypes expressed a dysfunctional program and lacked stem-like features among patients who lacked clinical benefit. Tracking tumor-reactive clonotypes over time, decay of antigen-reactive peripheral T cell clonotypes was associated with the emergence of progressive disease. Further, subclonal neoantigens previously targeted by infused T cells were subsequently absent within tumors at progression, suggesting potential adaptive resistance. Our findings suggest that targeting clonal antigens and circumventing dysfunctional states may be important for conferring clinical responses to TIL therapy.
Collapse
Affiliation(s)
- Chao Wang
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA.
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Jamie K Teer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Jiqiang Yao
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Dongliang Du
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Xiaoxian Liu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Zachary J Thompson
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Min Hsuan Wang
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Eric A Welsh
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Danish Memon
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-oncology, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Vladimir Makarov
- Center for Immunotherapy and Precision Immuno-oncology, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Carmen M Anadon
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
- Department of Integrative Immunobiology, Duke School of Medicine, Durham, NC, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Lamees Saeed
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Theresa A Boyle
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Bin Fang
- Proteomics & Metabolomics Core Facility, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - John M Koomen
- Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Cheryl Cox
- Immune and Cellular Therapy Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Ana M Landin
- Immune and Cellular Therapy Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Sean J Yoder
- Molecular Genomics Core Facility, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Sungjune Kim
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
- Department of Radiation Oncology, Immunology, Cancer Biology, Mayo Clinic Alix College of Medicine & Health Sciences, Jacksonville, FL, USA
| | - Dung-Tsa Chen
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Shari A Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
- Department of Integrative Immunobiology, Duke School of Medicine, Durham, NC, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Scott J Antonia
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Benjamin C Creelan
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA.
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA.
| |
Collapse
|
18
|
Liang Z, Tian F. Functional nucleic acid-based fluorescence imaging for tumor microenvironment monitoring: A review. Anal Chim Acta 2025; 1350:343794. [PMID: 40155176 DOI: 10.1016/j.aca.2025.343794] [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: 12/11/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND The tumor microenvironment (TME) refers to the complex ecological system surrounding tumor cells, which is intimately associated with regulating tumor cell growth, invasive behavior, and metastatic capacity. Hence, in situ imaging of related bioactivity with resolution in the TME is critical for early cancer detection and accurate diagnosis. In recent years, fluorescence imaging technology has become a widely used tool in TME research due to its non-invasive nature, high spatiotemporal resolution, and capability for real-time monitoring. Among these advancements, signal probes designed based on functional nucleic acids (FNAs) provide a promising and innovative toolkit for targeted imaging analysis of the TME. RESULTS This review provides a comprehensive discussion on the construction of FNA-based biosensors and their advancements in TME monitoring. In this review, we initially provide a systematic summary of the current targeting strategies of FNA-based biosensors for visual monitoring of the TME, focusing on targeting cell surface and extracellular matrix components. Subsequently, we further explore the application of FNA-based biosensors in monitoring the TME. These biosensors have successfully achieved the monitoring of key parameters, bioactive molecules and other tumor markers in the tumor microenvironment due to their excellent molecular recognition ability and high sensitivity. Finally, we discuss some of the challenges currently faced in the field. In response to these challenges, we propose potential research directions and look forward to the future development prospects of this field. SIGNIFICANCE Unlike previous reviews of biosensors based on FNAs for imaging tumor markers in the TME, this work is the first to review how such biosensors can be anchored in the TME. With continued efforts and advancements, we believe an increasing number of FNA-based fluorescence imaging probes will be utilized for TME imaging. This progress will significantly enhance our understanding of disease pathogenesis and progression, thereby offering substantial potential in biosensing and imaging analysis.
Collapse
Affiliation(s)
- Zuoxiang Liang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, 350122, PR China; Department of Epidemiology and Health Statistics & Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350122, PR China
| | - Fengyu Tian
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, 350122, PR China.
| |
Collapse
|
19
|
Garemilla SSS, Gampa SC, Garimella S. Role of the tumor microenvironment in cancer therapy: unveiling new targets to overcome drug resistance. Med Oncol 2025; 42:202. [PMID: 40332723 DOI: 10.1007/s12032-025-02754-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 04/28/2025] [Indexed: 05/08/2025]
Abstract
Cancer is a leading cause of death globally, with resistance to therapy representing a major obstacle to effective treatment. The tumor microenvironment (TME), comprising a complex network to cellular and non-cellular components including cancer-associated fibroblasts, immune cells, the extracellular matrix and region of hypoxia, is integral to cancer progression and therapeutic resistance. This review delves into the multifaceted interactions within the TME that contribute to tumor growth, survival and immune evasion. Key elements such as the role of cancer- associated fibroblasts in remodeling the extracellular matrix and promoting angiogenesis, the influence of immune cells such as tumor-associated macrophages in creating an immunosuppressive milieu and the impact of hypoxia conditions on metabolic adaptation and therapy resistance are thoroughly examined. This review evaluates current and emerging TME-targeted therapeutic strategies, including inhibitors of extracellular matrix components, modulators of immune cell activity and approached to alleviate hypoxia. Combination therapies that integrate TME-targeted agents with conventional treatments such as chemotherapy and immunotherapy are also discussed for their potential to enhance treatment efficacy and circumvent resistance mechanisms. By synthesising recent advances in TME research and therapeutic innovation, this paper aims to underscore the importance of TME in cancer therapy and highlight promising avenues for improving patient outcomes through targeted intervention.
Collapse
Affiliation(s)
| | - Siri Chandana Gampa
- Department of Life Sciences, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh, 530045, India
| | - Sireesha Garimella
- Department of Life Sciences, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh, 530045, India.
| |
Collapse
|
20
|
Liu S, Cao H, Wang Z, Zhu J, An X, Zhang L, Song Y. Single-cell transcriptomics reveals extracellular matrix remodeling and collagen dynamics during lactation in sheep mammary gland. Int J Biol Macromol 2025; 312:143669. [PMID: 40319976 DOI: 10.1016/j.ijbiomac.2025.143669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/13/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
The mammary gland is a dynamic organ with diverse cell populations that maintain glandular homeostasis, particularly during lactation. However, the cellular architecture and molecular mechanisms underlying lactational remodeling in the sheep mammary gland remain incompletely understood. Given similarities in mammary stromal structure, sheep serve as a valuable model for studying lactational changes relevant to the human breast, which experiences collagen loss and sagging during lactation. Utilizing single-cell transcriptomics (scRNA-seq), we mapped the sheep mammary gland's cellular landscape at postpartum days 60 and 150, identifying seven major cell types, including six distinct epithelial clusters. These clusters revealed differentiation among luminal progenitors, hormone-sensing, and myoepithelial cells across peak and late lactation stages. Transcriptomic analysis highlighted pivotal roles for epithelial integrity and ECM remodeling, with myoepithelial cells centrally involved in these processes. We observed significant collagen remodeling driven by fibroblast-epithelial crosstalk and ECM reorganization during late lactation. Comparative analysis with human mammary epithelial cells showed conserved basal and myoepithelial cell populations, while luminal cells diverged across species. This study provides insights into lactation biology and ECM remodeling, offering a framework to inform future studies on lactational adaptation and its implications for human health.
Collapse
Affiliation(s)
- Shujuan Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Heran Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Zhanhang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Junru Zhu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xiaopeng An
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Lei Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Yuxuan Song
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
21
|
Githaka JM, Kirschenman R, Patel N, Tripathi N, Wang J, Li L, Muranyi H, Pirayeshfard L, Montpetit R, Glubrecht DD, Lerner EP, Perry T, Danial NN, Nation PN, Godbout R, Goping IS. Multiple anti-tumor programs are activated by blocking BAD phosphorylation. Oncogene 2025:10.1038/s41388-025-03420-1. [PMID: 40316741 DOI: 10.1038/s41388-025-03420-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 05/04/2025]
Abstract
The Bcl-2 family member BAD is a candidate disease modulator because it stimulates apoptosis in a cell context basis and inhibits cell migration during normal mammary gland morphogenesis. This activity depends on 3 key regulatory serines (S75, 99, 118) in the unphosphorylated state. Given that developmental programs are often hijacked in cancer, we hypothesized that BAD would impede breast cancer progression. We generated breast cancer mouse models representing loss-of-function or phosphorylation deficient mutations (PyMT-Bad-/- and PyMT-Bad3SA/3SA, respectively). Preventing BAD phosphorylation significantly decreased breast cancer progression and metastasis. The knock-out phenocopied the control PyMT-Bad+/+ suggesting that phosphorylated BAD protein was inert. Thus, the BAD3SA mutation unmasked latent anti-tumor activity. Indeed, transcriptomics showed PyMT-Bad3SA/3SA activated multiple anti-tumor programs including apoptosis, inflammation, cellular differentiation, and diminished cell migration. This anti-tumor effect associated with clinical survival of breast cancer patients whose tumors had high levels of unphosphorylated BAD. Kinase screens identified ERK as the major BAD kinase in breast cells, and ERK inhibition impeded tumoroid invasion. Our data suggest that unphosphorylated BAD modulates anti-tumor pathways that contribute to excellent patient prognosis. Thus, targeting ERK to dephosphorylate BAD may be an exciting therapeutic opportunity in the future.
Collapse
Affiliation(s)
| | - Raven Kirschenman
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Namrata Patel
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Namita Tripathi
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Joy Wang
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Laiji Li
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Heather Muranyi
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | | | - Rachel Montpetit
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | | | - E Paul Lerner
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Troy Perry
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Nika N Danial
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - P Nick Nation
- Animal Pathology Services (APS) Ltd., Canmore, AB, Canada
| | - Roseline Godbout
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Ing Swie Goping
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.
- Department of Oncology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
22
|
Chao YL, Zhou KI, Forbes KK, Porrello A, Gentile GM, Zhu Y, Chack AC, John Mary DJS, Liu H, Cockman E, Edatt L, Goda GA, Zhao JJ, Abou Assi H, Wiedner HJ, Tsai Y, Wilkinson L, Van Swearingen AED, Carey LA, Giudice J, Dominguez D, Holley CL, Pecot CV. Snord67 promotes breast cancer metastasis by guiding U6 modification and modulating the splicing landscape. Nat Commun 2025; 16:4118. [PMID: 40316533 PMCID: PMC12048515 DOI: 10.1038/s41467-025-59406-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 04/22/2025] [Indexed: 05/04/2025] Open
Abstract
Previously considered "housekeeping" genes, small nucleolar RNAs (snoRNAs) are increasingly understood to have wide-ranging functions in cancer, yet their role in metastasis has been less well studied. Here, we identify the snoRNA Snord67 as a regulator of lymph node (LN) metastasis in breast cancer. Snord67 expression is enriched in LN metastases in an immune-competent mouse model of female breast cancer. In an orthotopic breast cancer model, loss of Snord67 decreases LN metastasis. In a model of lymphatic metastasis, Snord67 loss decreases LN tumor growth and distant metastases. In breast cancer cell lines, Snord67 knockout results in loss of targeted 2'-O-methylation on U6 small nuclear RNA, as well as widespread changes in splicing. Together, these results demonstrate that Snord67 regulates splicing and promotes the growth of LN metastases and subsequent spread to distant metastases. SnoRNA-guided modifications of the spliceosome and regulation of splicing may represent a potentially targetable pathway in cancer.
Collapse
Affiliation(s)
- Yvonne L Chao
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- Division of Hematology & Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Hematology & Oncology, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
- VA Pittsburgh Health System, Pittsburgh, PA, USA
| | - Katherine I Zhou
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kwame K Forbes
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alessandro Porrello
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabrielle M Gentile
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology (GMB), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yinzhou Zhu
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Aaron C Chack
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology (GMB), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dixcy J S John Mary
- Division of Hematology & Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Haizhou Liu
- Division of Hematology & Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eric Cockman
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Lincy Edatt
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Grant A Goda
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Justin J Zhao
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Hala Abou Assi
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Hannah J Wiedner
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology (GMB), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yihsuan Tsai
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Lily Wilkinson
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | | | - Lisa A Carey
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- Division of Hematology & Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jimena Giudice
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology (GMB), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daniel Dominguez
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA.
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Christopher L Holley
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | - Chad V Pecot
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA.
- Division of Hematology & Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
23
|
Chen Z, Xu L, Yuan Y, Zhang S, Xue R. Metabolic crosstalk between platelets and cancer: Mechanisms, functions, and therapeutic potential. Semin Cancer Biol 2025; 110:65-82. [PMID: 39954752 DOI: 10.1016/j.semcancer.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Platelets, traditionally regarded as passive mediators of hemostasis, are now recognized as pivotal regulators in the tumor microenvironment, establishing metabolic feedback loops with tumor and immune cells. Tumor-derived signals trigger platelet activation, which induces rapid metabolic reprogramming, particularly glycolysis, to support activation-dependent functions such as granule secretion, morphological changes, and aggregation. Beyond self-regulation, platelets influence the metabolic processes of adjacent cells. Through direct mitochondrial transfer, platelets reprogram tumor and immune cells, promoting oxidative phosphorylation. Additionally, platelet-derived cytokines, granules, and extracellular vesicles drive metabolic alterations in immune cells, fostering suppressive phenotypes that facilitate tumor progression. This review examines three critical aspects: (1) the distinctive metabolic features of platelets, particularly under tumor-induced activation; (2) the metabolic crosstalk between activated platelets and other cellular components; and (3) the therapeutic potential of targeting platelet metabolism to disrupt tumor-promoting networks. By elucidating platelet metabolism, this review highlights its essential role in tumor biology and its therapeutic implications.
Collapse
Affiliation(s)
- Zhixue Chen
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lin Xu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yejv Yuan
- The First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232001, China
| | - Si Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Ruyi Xue
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
24
|
Wu M, Yang H, Liu S, Jiang L, Liang T, Wang Y, Zhu M, Song X, Liu H, Shen J, Wang S, Zhu X, Qu CK, Cheng L, Jiang H, Ni F. Enhanced engraftment of human haematopoietic stem cells via mechanical remodelling mediated by the corticotropin-releasing hormone. Nat Biomed Eng 2025; 9:754-771. [PMID: 39715892 DOI: 10.1038/s41551-024-01316-1] [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: 09/27/2023] [Accepted: 11/01/2024] [Indexed: 12/25/2024]
Abstract
The engraftment of haematopoietic stem and progenitor cells (HSPCs), particularly in cord-blood transplants, remains challenging. Here we report the role of the corticotropin-releasing hormone (CRH) in enhancing the homing and engraftment of human-cord-blood HSPCs in bone marrow through mechanical remodelling. By using microfluidics, intravital two-photon imaging and long-term-engraftment assays, we show that treatment with CRH substantially enhances HSPC adhesion, motility and mechanical remodelling, ultimately leading to improved bone-marrow homing and engraftment in immunodeficient mice. CRH induces Ras homologue gene family member A (RhoA)-dependent nuclear translocation of the yes-associated protein (YAP), which upregulates the expression of genes encoding extracellular-matrix proteins (notably, thrombospondin-2 (THBS2)). This process guides the mechanical remodelling of HSPCs via modulation of the actin cytoskeleton and the extracellular matrix, with THBS2 interacting with the integrin αvβ3 and coordinating the nuclear translocation of YAP upon CRH/CRH-receptor-1 (CRH/CRHR1) signalling. Overall, the CRH/CRHR1/RhoA/YAP/THBS2/αvβ3 axis has a central role in modulating HSPC behaviour via a mechanical feedback loop involving THBS2, αvβ3, the actin cytoskeleton and YAP signalling. Our findings may suggest avenues for optimizing the transplantation of HSPCs.
Collapse
Affiliation(s)
- Mingming Wu
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Haoxiang Yang
- The CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, China
| | - Senquan Liu
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lai Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tingting Liang
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yan Wang
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mingming Zhu
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xian Song
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hao Liu
- The CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, China
| | - Jinghao Shen
- The CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, China
| | - Shuangzi Wang
- The CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, China
| | - Xiaoyu Zhu
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Cheng-Kui Qu
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Linzhao Cheng
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Hongyuan Jiang
- The CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, China.
| | - Fang Ni
- Department of Hematology, The First Affiliated Hospital of USTC, Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Blood and Cell Therapy and Anhui Provincial Key Laboratory of Blood Research and Applications, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Institute of Immunology, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
25
|
Li L, Wu J, Wu X, Li Z, Zhang X, Yan Z, Liang Y, Huang C, Qu S. Carbon Dot-Linked Hydrogel for TAMs Transform: Spatiotemporal Manipulation to Reshape Tumor Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2420068. [PMID: 40181663 PMCID: PMC12087752 DOI: 10.1002/adma.202420068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/20/2025] [Indexed: 04/05/2025]
Abstract
As one of the most crucial immune cells in the tumor microenvironment (TME), regulating tumor-associated macrophages (TAMs) is vital for enhancing antitumor immunity. Here, an injectable carbon dots (CDs)-linked egg white hydrogel was developed, termed TAMs Transform Factory (TTF-L-C), to spatiotemporally manipulate TAMs. The fabricated CDs significantly promoted macrophage migration. Notably, TTF-L-C achieved macrophage spatial enrichment through CDs-induced directional recruitment with molecular Ctnnd1 upregulation. Subsequently, the recruited macrophages were locoregionally reprogrammed within TTF-L-C, as well as blocking the upregulated PD-L1. Finally, through multi-stage regulation at spatial, cellular, and molecular levels, TTF-L-C released immune-activated M1 macrophages to the tumor site as it degraded. Moreover, TTF-L-C promoted dendritic cell (DCs) maturation and further boosted T cell activation, thereby reshaping the tumor-suppressive TME. Through peritumoral injection, TTF-L-C enhanced tumor immunotherapy in both subcutaneous and recurrent 4T1 tumor models with satisfactory biosafety. Therefore, TTF-L-C is proposed to become a safe and powerful platform for various biomedical applications.
Collapse
Affiliation(s)
- Lingyun Li
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Jun Wu
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Xue Wu
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Zhenjian Li
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Xianming Zhang
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Zekun Yan
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Yingqi Liang
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Caishi Huang
- Faculty of Health ScienceUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Songnan Qu
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
- Department of Physics and ChemistryFaculty of Science and Technology University of MacauMacao SAR999078P. R. China
- MOE Frontier Science Centre for Precision Oncology University of MacauMacao SAR999078P. R. China
- Zhuhai UM Science and Technology Research InstituteZhuhai519031P. R. China
| |
Collapse
|
26
|
Li Y, Sun Y, Yu K, Li Z, Miao H, Xiao W. Keratin: A potential driver of tumor metastasis. Int J Biol Macromol 2025; 307:141752. [PMID: 40049479 DOI: 10.1016/j.ijbiomac.2025.141752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
Keratins, as essential components of intermediate filaments in epithelial cells, play a crucial role in maintaining cell structure and function. In various malignant epithelial tumors, abnormal keratin expression is frequently observed and serves not only as a diagnostic marker but also closely correlates with tumor progression. Extensive research has demonstrated that keratins are pivotal in multiple stages of tumor metastasis, including responding to mechanical forces, evading the immune system, reprogramming metabolism, promoting angiogenesis, and resisting apoptosis. Here we emphasize that keratins significantly enhance the migratory and invasive capabilities of tumor cells, making them critical drivers of tumor metastasis. These findings highlight the importance of targeting keratins as a strategic approach to combat tumor metastasis, thereby advancing our understanding of their role in cancer progression and offering new therapeutic opportunities.
Collapse
Affiliation(s)
- Yuening Li
- Army Medical University, Chongqing, China
| | - Yiming Sun
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Kun Yu
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Zhixi Li
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China.
| | - Hongming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China; Jinfeng Laboratory, Chongqing, China.
| | - Weidong Xiao
- Department of General Surgery, the Second Affiliated Hospital of Army Medical University, Chongqing, China.
| |
Collapse
|
27
|
Li K, He Y, Jin X, Jin K, Qian J. Reproducible extracellular matrices for tumor organoid culture: challenges and opportunities. J Transl Med 2025; 23:497. [PMID: 40312683 PMCID: PMC12044958 DOI: 10.1186/s12967-025-06349-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: 01/03/2025] [Accepted: 03/03/2025] [Indexed: 05/03/2025] Open
Abstract
Tumor organoid models have emerged as valuable 3D in vitro systems to study cancer behavior in a physiologically relevant environment. The composition and architecture of the extracellular matrix (ECM) play critical roles in tumor organoid culture by influencing the tumor microenvironment and tumor behavior. Traditional matrices such as Matrigel and collagen, have been widely used, but their batch-to-batch variability and limited tunability hinder their reproducibility and broader applications. To address these challenges, researchers have turned to synthetic/engineered matrices and biopolymer-based matrices, which offer precise tunability, reproducibility, and chemically defined compositions. However, these matrices also present challenges of their own. In this review, we explore the significance of ECMs in tumor organoid culture, discuss the limitations of commonly used matrices, and highlight recent advancements in engineered/synthetic matrices for improved tumor organoid modeling.
Collapse
Affiliation(s)
- Kan Li
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yibo He
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, 310006, China
- Department of Breast Surgery, Affiliated Hangzhou First People'S Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, China
| | - Xue Jin
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People'S Hospital (Affiliated People'S Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Ketao Jin
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310003, China.
| | - Jun Qian
- Department of Colorectal Surgery, Xinchang People'S Hospital, Affiliated Xinchang Hosptial, Wenzhou Medical University, Xinchang, Zhejiang, 312500, China.
| |
Collapse
|
28
|
Romoli J, Chiodelli P, Signoroni PB, Vertua E, Ferrari C, Giuzzi E, Paini A, Scalvini E, Papait A, Stefani FR, Silini AR, Parolini O. Modeling Stromal Cells Inside the Tumor Microenvironment of Ovarian Cancer: In Vitro Generation of Cancer-Associated Fibroblast-Like Cells and Their Impact in a 3D Model. MedComm (Beijing) 2025; 6:e70172. [PMID: 40255916 PMCID: PMC12006666 DOI: 10.1002/mco2.70172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 01/31/2025] [Accepted: 02/13/2025] [Indexed: 04/22/2025] Open
Abstract
The tumor microenvironment (TME) is the combination of cells and factors that promotes tumor progression, and cancer-associated fibroblasts (CAFs) are a key component within TME. CAF originates from various stromal cells and is activated by factors such as transforming growth factor-beta (TGF-β) secreted by tumor cells, favoring chemoresistance and metastasis. Recent publications have underlined plasticity and heterogeneity and their strong contribution to the reactive stroma within the TME. Our study aimed to replicate the TME's structure by creating a 3D in vitro model of ovarian cancer (OC). By incorporating diverse tumor and stromal cells, we simulated a physiologically relevant environment for studying CAF-like cell behavior within tumor spheroids in a context-dependent manner. CAF-like cells were generated by exposing human dermal fibroblasts to OC cell line conditioned media in the presence or absence of TGF-β. Herein, we found that different stimuli induce the generation of heterogeneous populations of CAF-like cells. Notably, we observed the ability of CAF-like cells to shape the intratumoral architecture and to contribute to functional changes in tumor cell behavior. This study highlights the importance of precise assessment of CAF for potential therapeutic interventions and further provides a reliable model for investigating novel therapeutic targets in OC.
Collapse
Affiliation(s)
- Jacopo Romoli
- Department of Life Science and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
| | - Paola Chiodelli
- Department of Life Science and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
| | | | - Elsa Vertua
- Centro di Ricerca E. MenniFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Clarissa Ferrari
- Research and Clinical Trials UnitFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Elisabetta Giuzzi
- Centro di Ricerca E. MenniFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Alice Paini
- Centro di Ricerca E. MenniFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Elisa Scalvini
- Centro di Ricerca E. MenniFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Andrea Papait
- Department of Life Science and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCSRomeItaly
| | | | | | - Ornella Parolini
- Department of Life Science and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCSRomeItaly
| |
Collapse
|
29
|
He C, Zhou Z, Yang Y, Zhu S, Wang H, Teng L. FERMT2 drives anoikis resistance and peritoneal metastasis by enhancing extracellular matrix deposition in gastric cancer. Gastric Cancer 2025; 28:409-421. [PMID: 40024947 PMCID: PMC11993459 DOI: 10.1007/s10120-025-01602-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/15/2025] [Indexed: 03/04/2025]
Abstract
Peritoneal metastasis is a critical step in the progression of gastric cancer (GC), yet its underlying mechanisms remain poorly understood. Here, we identify FERMT2, a member of the Kindlin protein family, as a key regulator of anoikis resistance (AR) and peritoneal metastasis in GC. FERMT2 expression increases in a suspension-time-dependent manner and is associated with higher pathological grade, advanced clinical stage, and poorer prognosis. Functional studies in vitro and in vivo demonstrate that FERMT2 promotes AR and facilitates peritoneal metastasis. Mechanistically, FERMT2 suppresses the ubiquitination of SOX2, thereby enhancing its stability and up-regulating FN1 transcription. Furthermore, we report that TGFβ-RI expression also increases in a suspension-time-dependent manner, forming a positive feedback loop with FERMT2 via TGFβ-1/TGFβ-RI signaling. This feedback loop drives extracellular fibronectin matrix deposition, strengthens cell-matrix interactions, and supports AR. These findings establish FERMT2 as a pivotal mediator of peritoneal metastasis in GC, offering insights into its potential as a therapeutic target.
Collapse
Affiliation(s)
- Chao He
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zheng Zhou
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Yang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Songting Zhu
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiyong Wang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lisong Teng
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
30
|
Akanda MR, Lubaba U, Rahman MK, Islam A, Akter M, Islam MS, Uddin MN, Park BY. Mechanistic role of stromal cancer-associated fibroblasts in tumorigenesis and brain metastasis: Highlighting drug resistance and targeted therapy. Pathol Res Pract 2025; 269:155918. [PMID: 40120401 DOI: 10.1016/j.prp.2025.155918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 03/09/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Brain metastases remain a major clinical challenge due to their high resistance to conventional and targeted therapies. Cancer-associated fibroblasts are the most common cellular component of the brain metastases tumor microenvironment. They significantly impact the tumor microenvironment because they promote cancer cell invasion, enhance metastasis, boost immune evasion, and contribute to drug resistance. We searched the PubMed and Google Scholar databases and included 99 studies to summarize the present review. Based on the searched articles, the present review emphasizes that biomarkers including PDGFR-β, α-SMA, and collagen I can identify metastatic brain cancer-associated fibroblasts, which lead to a poor prognosis and recurrence. In addition, cancer-associated fibroblasts can cause resistance to therapy by modifying the extracellular matrix (e.g., collagen I, fibronectin), secreting growth factors (e.g., TGF-β, HGF, IL-6), causing immunological evasion (e.g., Tregs, MDSCs), secreting exosomes (e.g., miRNAs), metabolic reprogramming, stemness induction, and plasticity. We also describe the molecular mechanisms by which cancer-associated fibroblasts confer drug resistance in brain metastases, such as extracellular matrix restoration, immunological evasion, metabolic reprogramming, etc. We also cover prospective therapeutic options for overcoming medication resistance, such as cancer-associated fibroblasts depletion, paracrine signaling blockage, metabolic inhibitors, and cancer-associated fibroblasts-targeted immunotherapies. Targeting cancer-associated fibroblasts in addition to existing medications may improve cancer treatment efficacy and survival rates for individuals with brain metastases. However, more research is required to better understand their role in metastatic brain tumors.
Collapse
Affiliation(s)
- Md Rashedunnabi Akanda
- Department of Pharmacology and Toxicology, Sylhet Agricultural University, Sylhet, Bangladesh.
| | - Umme Lubaba
- Department of Pharmacology and Toxicology, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Md Khalesur Rahman
- Department of Microbiology and Immunology, East Carolina University, Greenville, NC, USA; Department of Microbiology, Hajee Mohammad Danesh Science & Technology University, Dinajpur, Bangladesh
| | - Anowarul Islam
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, Australia; Epilepsy Research Group, Australian Centre for Precision Health, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Momota Akter
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Md Sadikul Islam
- Vascular Biology Center, Medical College of Georgia, Augusta University, GA, USA
| | - Md Nazim Uddin
- Department of Livestock Production and Management, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Byung-Yong Park
- Institute of Animal Transplantation, College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea
| |
Collapse
|
31
|
Marjan T, Lafuente-Gómez N, Rampal A, Mooney DJ, Peyton SR, Qazi TH. Cell-Instructive Biomaterials with Native-Like Biochemical Complexity. Annu Rev Biomed Eng 2025; 27:185-209. [PMID: 39874600 PMCID: PMC12045723 DOI: 10.1146/annurev-bioeng-120823-020209] [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] [Indexed: 01/30/2025]
Abstract
Biochemical signals in native tissue microenvironments instruct cell behavior during many biological processes ranging from developmental morphogenesis and tissue regeneration to tumor metastasis and disease progression. The detection and characterization of these signals using spatial and highly resolved quantitative methods have revealed their existence as matricellular proteins in the matrisome, some of which are bound to the extracellular matrix while others are freely diffusing. Including these biochemical signals in engineered biomaterials can impart enhanced functionality and native-like complexity, ultimately benefiting efforts to understand, model, and treat various diseases. In this review, we discuss advances in characterizing, mimicking, and harnessing biochemical signals in developing advanced engineered biomaterials. An overview of the diverse forms in which these biochemical signals exist and their effects on intracellular signal transduction is also provided. Finally, we highlight the application of biochemically complex biomaterials in the three broadly defined areas of tissue regeneration, immunoengineering, and organoid morphogenesis.
Collapse
Affiliation(s)
- Tuba Marjan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| | - Nuria Lafuente-Gómez
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA;
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | - Akaansha Rampal
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA;
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | - Shelly R Peyton
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA;
| | - Taimoor H Qazi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| |
Collapse
|
32
|
Oh JM, Park Y, Lee J, Shen K. Microfabricated Organ-Specific Models of Tumor Microenvironments. Annu Rev Biomed Eng 2025; 27:307-333. [PMID: 40310890 DOI: 10.1146/annurev-bioeng-110222-103522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Despite the advances in detection, diagnosis, and treatments, cancer remains a lethal disease, claiming the lives of more than 600,000 people in the United States alone in 2024. To accelerate the development of new therapeutic strategies with improved responses, significant efforts have been made to develop microfabricated in vitro models of tumor microenvironments (TMEs) that address the limitations of animal-based cancer models. These models incorporate several advanced tissue engineering techniques to better reflect the organ- and patient-specific TMEs. Additionally, microfabricated models integrated with next-generation single-cell omics technologies provide unprecedented insights into patient's cellular and molecular heterogeneity and complexity. This review provides an overview of the recent understanding of cancer development and outlines the key TME elements that can be captured in microfabricated models to enhance their physiological relevance. We highlight the recent advances in microfabricated cancer models that reflect the unique characteristics of their organs of origin or sites of dissemination.
Collapse
Affiliation(s)
- Jeong Min Oh
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA;
| | - Yongkuk Park
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA;
| | - Jungwoo Lee
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA;
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| | - Keyue Shen
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA;
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
33
|
Tang F, Zhu Y, Shen J, Yuan B, He X, Tian Y, Weng L, Sun L. CD44 + cells enhance pro-tumor stroma in the spatial landscape of colorectal cancer leading edge. Br J Cancer 2025; 132:703-715. [PMID: 40069574 PMCID: PMC11997037 DOI: 10.1038/s41416-025-02968-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 02/04/2025] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND The heterogeneity of tumors significantly impacts on colorectal cancer (CRC) progression. However, the influence of this heterogeneity on the spatial architecture of CRC remains largely unknown. METHODS Spatial transcriptomic (ST) analysis of AOM/DSS-induced colorectal cancer (CRC), integrated with single-cell RNA sequencing, generated a comprehensive spatial atlas of CRC. Pseudotime trajectory, stemness evaluation, and cell-cell communication analyses explored how CD44+ tumor cells at the leading edge remodel the tumor microenvironment (TME). In vitro experiments and immunofluorescence staining of clinical samples validated pleiotrophin (PTN) signaling in promoting cancer-associated fibroblasts (CAFs) phenotypic transition and CRC progression. RESULTS Our findings revealed a distinctive layered ring-like structure within CRC tissues, where CD44+ tumor cells exhibiting high stemness were positioned at the tumor's leading edge. Inflammatory CAFs (iCAFs)-like, myofibroblastic CAFs (myCAFs)-like cells and pro-tumorigenic neutrophils primarily located at the tumor edge, in proximity to CD44+ tumor cells. CD44+ tumor cells then triggered the phenotypic transition of CAFs into iCAF-like and myCAF-like cells through PTN signaling. CONCLUSIONS Our results provide distinctive insights into how tumor heterogeneity reshapes the TME at the leading edge of tumor, thereby promoting CRC progression.
Collapse
Affiliation(s)
- Feiyu Tang
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, China
- Center for Biotherapy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yongwei Zhu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Jia Shen
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, China
| | - Bowen Yuan
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiang He
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, China
| | - Yuxi Tian
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Liang Weng
- Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, China.
| | - Lunquan Sun
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, China.
- FuRong Laboratory, Changsha, China.
| |
Collapse
|
34
|
Li W, Wu Y, Zhang Y, Gao W, Li X, Luo H, Lu M, Liu Z, Luo A. Halofuginone Disrupted Collagen Deposition via mTOR-eIF2α-ATF4 Axis to Enhance Chemosensitivity in Ovarian Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416523. [PMID: 40126173 DOI: 10.1002/advs.202416523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/14/2025] [Indexed: 03/25/2025]
Abstract
The interplay between cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM) mediates progress, metastasis, and therapy resistance. However, strategy of targeting ECM remodeling to enhance chemosensitivity in ovarian cancer remains elusive. Here, a 22-gene matrisome signature predicts chemotherapy response and survival in ovarian cancer. The dense, collagen-rich ECM secreted by CAFs harbors more M2 tumor-associated macrophages (TAMs) than the looser ECM based on single cell RNA-seq (scRNA-seq) of ovarian cancer, suggesting the promising approach of targeting collagen to remodel ECM. An integrated analysis identifies collagen type I alpha 1 chain (COL1A1) as a major component of the ECM that contributes to chemoresistance and poor prognosis, highlighting its potential as a therapeutic target. Halofuginone (HF), a clinically active derivative of febrifugine, is identified as a COL1A1-targeting natural compound by screening the Encyclopedia of Traditional Chinese Medicine (ETCM). Mechanistically, HF inhibits COL1A1 production via the mTOR-eIF2α-ATF4 axis in CAFs. Notably, HF disrupts collagen deposition and promotes CD8+ T cell infiltration, partially via M2-M1 macrophage polarization to enhance chemosensitivity. Overall, the findings suggest that HF combined with chemotherapy is a promising and effective treatment for ovarian cancer.
Collapse
Affiliation(s)
- Wenxin Li
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Nanli Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Yenan Wu
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Nanli Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Yanan Zhang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, 38 Xueyuan Rd, Haidian District, Beijing, 100191, China
| | - Wenyan Gao
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Nanli Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Xin Li
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Nanli Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Haixia Luo
- Department of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Nanli Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Mengmeng Lu
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Nanli Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Zhihua Liu
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Nanli Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Aiping Luo
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Nanli Panjiayuan, Chaoyang District, Beijing, 100021, China
| |
Collapse
|
35
|
Chen R, Zhang R, Ke F, Guo X, Zeng F, Liu Q. Mechanisms of breast cancer metastasis: the role of extracellular matrix. Mol Cell Biochem 2025; 480:2771-2796. [PMID: 39652293 DOI: 10.1007/s11010-024-05175-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/23/2024] [Indexed: 05/03/2025]
Abstract
The components of the extracellular matrix (ECM) are dynamic, and they mediate mechanical signals that modulate cellular behaviors. Disruption of the ECM can induce the migration and invasion of cancer cells via specific signaling pathways and cytokines. Metastasis is a leading cause of high mortality in malignancies, and early intervention can improve survival rates. However, breast cancer is frequently diagnosed subsequent to metastasis, resulting in poor prognosis and distant metastasis poses substantial hurdles in therapy. In breast cancer, there is notable tissue remodeling of ECM proteins, with several identified as essential components for metastasis. Moreover, specific ECM molecules, receptors, enzymes, and various signaling pathways play crucial roles in breast cancer metastasis, drug treatment, and resistance. The in-depth consideration of these elements could provide potential therapeutic targets to enhance the survival rates and quality of life for breast cancer patients. This review explores the mechanisms by which alterations in the ECM contribute to breast cancer metastasis and discusses current clinical applications targeting ECM in breast cancer treatment, offering valuable perspectives for future ECM-based therapies.
Collapse
Affiliation(s)
- Rui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Ranqi Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Famin Ke
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiurong Guo
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
36
|
Chang CC, Chang CB, Shen CH, Lee MY, Jou YC, Tung CL, Lai WH, Hung CF, Wang M, Lai YY, Chen PC, Wu SF. Immunosuppression of Tumor-Derived Factors Modulated Neutrophils in Upper Tract Urothelial Carcinoma Through Upregulation of Arginase-1 via ApoA1-STAT3 Axis. Cells 2025; 14:660. [PMID: 40358184 PMCID: PMC12072159 DOI: 10.3390/cells14090660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Upper tract urothelial carcinoma (UTUC) presents aggressive features and a tumor microenvironment with T cell depletion. However, the role of tumor-associated neutrophils in UTUC remains unclear. This study aimed to investigate how UTUC tumor-derived factors modulate neutrophils and their impact on T cell immune responses. Our findings demonstrate that UTUC secreted tumor-derived factors, with apolipoprotein A1 (Apo-A1) being the predominant factor, which upregulated arginase-1 expression in neutrophils. STAT3 activation was responsible for the upregulation of arginase-1 in neutrophils. Blocking the interactions between Apo-A1 and its receptors reduced arginase-1 expression in neutrophils treated with tumor tissue culture supernatant (TTCS). Moreover, both CD4+ T and CD8+ T cell proliferation were inhibited by neutrophils treated with Apo-A1 or TTCS. Importantly, blocking Apo-A1 signaling in neutrophils reversed the inhibitory effects on T cells. In UTUC patients, the neutrophil-to-lymphocyte ratio was higher than that in healthy subjects. The expression of arginase-1 in neutrophils and the level of Apo-A1 within UTUC tumors were negatively correlated with tumor-infiltrating CD4+ T cells. Additionally, neutrophils from UTUC patients showed increased expression of arginase-1 and exhibited inhibitory effects of T cell functions. These findings suggest that UTUC orchestrates an immune-suppressive microenvironment through Apo-A1-mediated upregulation of arginase-1 in neutrophils, ultimately leading to the inhibition of T cell proliferation.
Collapse
Affiliation(s)
- Chih-Chia Chang
- Department of Radiation Therapy and Oncology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan;
| | - Chia-Bin Chang
- Department of Urology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan; (C.-B.C.); (C.-H.S.); (W.-H.L.); (C.-F.H.)
| | - Cheng-Huang Shen
- Department of Urology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan; (C.-B.C.); (C.-H.S.); (W.-H.L.); (C.-F.H.)
| | - Ming-Yang Lee
- Department of Hematology and Oncology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan;
| | - Yeong-Chin Jou
- Department of Urology, St. Martin De Porres Hospital, Chiayi 60069, Taiwan;
| | - Chun-Liang Tung
- Department of Pathology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan;
| | - Wei-Hong Lai
- Department of Urology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan; (C.-B.C.); (C.-H.S.); (W.-H.L.); (C.-F.H.)
| | - Chi-Feng Hung
- Department of Urology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan; (C.-B.C.); (C.-H.S.); (W.-H.L.); (C.-F.H.)
| | - Meilin Wang
- Department of Microbiology and Immunology, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan;
| | - Ya-Yan Lai
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 60004, Taiwan;
| | - Pi-Che Chen
- Department of Urology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan; (C.-B.C.); (C.-H.S.); (W.-H.L.); (C.-F.H.)
| | - Shu-Fen Wu
- Department of Biomedical Sciences, and Epigenomics Human Disease Research Center, National Chung Cheng University, Chiayi 62102, Taiwan
| |
Collapse
|
37
|
Finger AM, Hendley AM, Figueroa D, Gonzalez H, Weaver VM. Tissue mechanics in tumor heterogeneity and aggression. Trends Cancer 2025:S2405-8033(25)00096-2. [PMID: 40307158 DOI: 10.1016/j.trecan.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 03/10/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Tumorigenesis ensues within a heterogeneous tissue microenvironment that promotes malignant transformation, metastasis and treatment resistance. A major feature of the tumor microenvironment is the heterogeneous population of cancer-associated fibroblasts and myeloid cells that stiffen the extracellular matrix. The heterogeneously stiffened extracellular matrix in turn activates cellular mechanotransduction and creates a hypoxic and metabolically hostile microenvironment. The stiffened extracellular matrix and elevated mechanosignaling also drive tumor aggression by fostering tumor cell growth, survival, and invasion, compromising antitumor immunity, expanding cancer stem cell frequency, and increasing mutational burden, which promote intratumor heterogeneity. Delineating the molecular mechanisms whereby tissue mechanics regulate these phenotypes should help to clarify the basis for tumor heterogeneity and cancer aggression and identify novel therapeutic targets that could improve patient outcome. Here, we discuss the role of the extracellular matrix in driving cancer aggression through its impact on tumor heterogeneity.
Collapse
Affiliation(s)
- Anna-Marie Finger
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA 94143; Current address: Liver Disease Research, Global Drug Discovery, Novo Nordisk A/S, Malov, Denmark
| | - Audrey Marie Hendley
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA 94143
| | - Diego Figueroa
- Department of Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Hugo Gonzalez
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA 94143; Current address: Laboratory of Tumor Microenvironment and Metastasis, Centro Ciencia & Vida, Santiago, Chile
| | - Valerie Marie Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA 94143; Department of Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
38
|
Chen J, Wang S, Ding Y, Xu D, Zheng S. Radiotherapy-induced alterations in tumor microenvironment: metabolism and immunity. Front Cell Dev Biol 2025; 13:1568634. [PMID: 40356601 PMCID: PMC12066526 DOI: 10.3389/fcell.2025.1568634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Tumor metabolism plays a pivotal role in shaping immune responses within the tumor microenvironment influencing tumor progression, immune evasion, and the efficacy of cancer therapies. Radiotherapy has been shown to impact both tumor metabolism and immune modulation, often inducing immune activation through damage-associated molecular patterns and the STING pathway. In this study, we analyse the particular characteristics of the tumour metabolic microenvironment and its effect on the immune microenvironment. We also review the changes in the metabolic and immune microenvironment that are induced by radiotherapy, with a focus on metabolic sensitisation to the effects of radiotherapy. Our aim is to contribute to the development of research ideas in the field of radiotherapy metabolic-immunological studies.
Collapse
Affiliation(s)
- Jinpeng Chen
- Department of General Surgery, Zhongda Hospital Southeast University, Nanjing, Jiangsu, China
- Southeast University Medical School, Nanjing, Jiangsu, China
| | - Sheng Wang
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, China
| | - Yue Ding
- Department of General Surgery, Zhongda Hospital Southeast University, Nanjing, Jiangsu, China
- Southeast University Medical School, Nanjing, Jiangsu, China
| | - Duo Xu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shiya Zheng
- Southeast University Medical School, Nanjing, Jiangsu, China
- Department of Oncology, Southeast University, Zhongda Hospital Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
39
|
Adlakha YK, Chhabra R. The human microbiome: redefining cancer pathogenesis and therapy. Cancer Cell Int 2025; 25:165. [PMID: 40296128 PMCID: PMC12039184 DOI: 10.1186/s12935-025-03787-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
The human microbiome has always been an important determinant of health and recently, its role has also been described in cancer. The altered microbiome could aid cancer progression, modulate chemoresistance and significantly alter drug efficacy. The broad implications of microbes in cancer have prompted researchers to investigate the microbe-cancer axis and identify whether modifying the microbiome could sensitize cancer cells for therapy and improve the survival outcome of cancer patients. The preclinical data has shown that enhancing the number of specific microbial species could restore the patients' response to cancer drugs and the microbial biomarkers may play a vital role in cancer diagnostics. The elucidation of detailed interactions of the human microbiota with cancer would not only help identify the novel drug targets but would also enhance the efficacy of existing drugs. The field exploring the emerging roles of microbiome in cancer is at a nascent stage and an in-depth scientific perspective on this topic would make it more accessible to a wider audience. In this review, we discuss the scientific evidence connecting the human microbiome to the origin and progression of cancer. We also discuss the potential mechanisms by which microbiota affects initiation of cancer, metastasis and chemoresistance. We highlight the significance of the microbiome in therapeutic outcome and evaluate the potential of microbe-based cancer therapy.
Collapse
Affiliation(s)
- Yogita K Adlakha
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Sector-125, Noida, Uttar Pradesh, 201303, India.
| | - Ravindresh Chhabra
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India.
| |
Collapse
|
40
|
Vasu N, Perumal T, Seetharaman PK, Sivapunniyam A, Anthoni G, Ramalingam KR, Rajamohan V, Thangaiyan S. Aegle marmelos-mediated zinc oxide nanoparticles for mosquito control, cancer therapy, and antibacterial applications. Bioprocess Biosyst Eng 2025:10.1007/s00449-025-03162-0. [PMID: 40293515 DOI: 10.1007/s00449-025-03162-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/27/2025] [Indexed: 04/30/2025]
Abstract
The study investigated the synthesis and characterization of zinc oxide nanoparticles (Am-ZnO-NPs) using Aegle marmelos leaves and their mosquitocidal, antioxidant, and anticancer efficacy. Am-ZnO-NPs were synthesized via combustion method under optimized conditions, including 5% plant extract, temperature of 70 °C, a contact time of 60 min, and pH of 10. The resulting nanoparticles were characterized using advanced analytical techniques. The Am-CuO-NPs were hexagonal wurtzite in shape and had an average particle size of 37.44 nm. The Am-CuO-NPs are highly stable with a surface charge of - 26.77 mV. Gas chromatography-mass spectrometry (GC-MS) analysis identified several compounds with insecticidal, antioxidant, and anticancer properties, including hexadecenoic acid, ethyl ester (25.61%), 9,12-octadecadienoic acid, ethyl ester (12.64%), and 9,12,15-octadecatrienoic acid, ethyl ester (6.31%). The aqueous extract of A. marmelos (100 µg/mL) demonstrated the highest larvicidal (57.33%) and pupicidal (42.67%) activities compared to other solvent extracts. In contrast, Am-ZnO-NPs exhibited relatively higher larvicidal activity (86.67%) and pupicidal (56.00%) at 10 µg/mL. The nanoparticles also inhibited the activities of acetylcholinesterase (AChE) and carboxylesterases and reduced the total protein (TP) levels in Ae. aegypti larvae. Further, both the aqueous leaf extract and Am-ZnO-NPs were effective in scavenging DPPH free radicals, achieving 76% and 94% inhibition, respectively, at a concentration of 125 µg/mL. Am-ZnO-NPs also exhibited cytotoxicity, induced cell cycle arrest, enhanced cell adhesion, and promoted apoptosis in L-132 lung cancer cell lines. Further, the Am-ZnO-NPs display potent antibacterial activity against clinical pathogens. These results highlight the role of phytochemicals in A. marmelos leaves for their effective biological activities.
Collapse
Affiliation(s)
- Nandhini Vasu
- Research Department of Zoology, Kunthavai Naacchiyaar Government Arts College for Women (A), (Affiliated By Bharathidasan University), Thanjavur, Tamil Nadu, 613007, India
| | - Thangamathi Perumal
- Research Department of Zoology, Kunthavai Naacchiyaar Government Arts College for Women (A), (Affiliated By Bharathidasan University), Thanjavur, Tamil Nadu, 613007, India
| | - Prabu Kumar Seetharaman
- Center for Applied Research, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, Tamil Nadu, 602105, India
- Laboratory of Functional Molecules and Materials, School of Physics and Optoelectronic Engineering, Shandong University of Technology, Xincun West Road 266, Zibo, 255000, China
| | - Ananth Sivapunniyam
- Center for Applied Research, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, Tamil Nadu, 602105, India.
| | - Gnanasoundari Anthoni
- Department of Hospital Administration, Queens College of Arts and Science for Women, Punalkulam, Pudukkottai, Tamil Nadu, India
| | - Karthik Raja Ramalingam
- Center for Applied Research, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, Tamil Nadu, 602105, India
| | - Vahitha Rajamohan
- Department of Zoology, M.V. Muthiah Government College (W), Dindigul, Tamil Nadu, 624001, India
| | - Sharmiladevi Thangaiyan
- Research Department of Zoology, Kunthavai Naacchiyaar Government Arts College for Women (A), (Affiliated By Bharathidasan University), Thanjavur, Tamil Nadu, 613007, India
| |
Collapse
|
41
|
Ding X, Liang Y, Zhou S, Wu Y, Sałata P, Mikolajczk-Martinez A, Khosrawipour V, Zhang Z. Targeting tumor extracellular matrix with nanoparticles to circumvent therapeutic resistance. J Control Release 2025; 383:113786. [PMID: 40306575 DOI: 10.1016/j.jconrel.2025.113786] [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: 03/07/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
Each stage of tumor development is intrinsically linked to the tumor microenvironment (TME), wherein the extracellular matrix (ECM) serves as a vital and abundant component in tumor tissues. The ECM is a non-cellular, three-dimensional macromolecular network scaffold that provides structural support to cells, stores bioactive molecules, and mediates signaling pathways through specific binding to cell surface receptors. Moreover, the ECM in tumor tissues plays a crucial role in impeding drug diffusion and resisting apoptosis induced by conventional anti-cancer therapies that primarily target cancer cells. Therefore, directing attentions towards the tumor ECM can facilitate the identification of novel targets and the development of new therapies. This review aims to summarize the composition, structure, remodeling, and function of tumor ECM, its association with drug resistance, and current targeting strategies, with a specific emphasis on nanoparticles (NPs).
Collapse
Affiliation(s)
- Xinyue Ding
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Yiyu Liang
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Siyuan Zhou
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Yao Wu
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Patricia Sałata
- Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | | | | | - Zhiwen Zhang
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China.
| |
Collapse
|
42
|
Rodella G, Préat V, Gallez B, Malfanti A. Design Strategies for Hyaluronic Acid-based Drug Delivery Systems in Cancer Immunotherapy. J Control Release 2025; 383:113784. [PMID: 40294800 DOI: 10.1016/j.jconrel.2025.113784] [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: 03/25/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
Despite its robust therapeutic potential, cancer immunotherapy has provided little progress towards improved survival rates for patients bearing immunologically refractory tumors. The implementation of advanced drug delivery systems represents a powerful means of improving cancer immunotherapy by relieving immunosuppression and promoting immune response; however, the overall impact of these systems on immunotherapy currently remains modest. Hyaluronic acid represents a widely used polymer in drug delivery; meanwhile, recent studies linking hyaluronic acid to the immune system make this polymer an attractive component in the design of next-generation cancer immunotherapies. Herein, we review our current understanding of the immunological properties of hyaluronic acid and discuss them in the context of bioactive functions and immune-related interactions with receptors, immune, and cancer cells. We analyze the potential of hyaluronic acid as a component in advanced drug delivery systems, highlighting strategies for the design of more effective vaccines and cancer chemo-immunotherapies. Finally, we discuss critical considerations to facilitate design and clinical translation to overcome existing challenges and maximize therapeutic potential.
Collapse
Affiliation(s)
- Giulia Rodella
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium; UCLouvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance, Avenue Mounier 73 B1.73.08, 1200 Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium
| | - Bernard Gallez
- UCLouvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance, Avenue Mounier 73 B1.73.08, 1200 Brussels, Belgium.
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium; Departement of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo, 5, 35131 Padova, Italy.
| |
Collapse
|
43
|
Cunningham C, Sun B. Representation of high-dimensional cell morphology and morphodynamics in 2D latent space. Phys Biol 2025; 22:10.1088/1478-3975/adcd37. [PMID: 40233771 PMCID: PMC12083545 DOI: 10.1088/1478-3975/adcd37] [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: 12/07/2024] [Accepted: 04/15/2025] [Indexed: 04/17/2025]
Abstract
The morphology and morphodynamics of cells as important biomarkers of the cellular state are widely appreciated in both fundamental research and clinical applications. Quantification of cell morphology often requires a large number of geometric measures that form a high-dimensional feature vector. This mathematical representation creates barriers to communicating, interpreting, and visualizing data. Here, we develop a deep learning-based algorithm to project 13-dimensional (13D) morphological feature vectors into 2-dimensional (2D) morphological latent space (MLS). We show that the projection has less than 5% information loss and separates the different migration phenotypes of metastatic breast cancer cells. Using the projection, we demonstrate the phenotype-dependent motility of breast cancer cells in the 3D extracellular matrix, and the continuous cell state change upon drug treatment. We also find that dynamics in the 2D MLS quantitatively agrees with the morphodynamics of cells in the 13D feature space, preserving the diffusive power and the Lyapunov exponent of cell shape fluctuations even though the dimensional reduction projection is highly nonlinear. Our results suggest that MLS is a powerful tool to represent and understand the cell morphology and morphodynamics.
Collapse
Affiliation(s)
- Christian Cunningham
- Department of Physics, Oregon State University, Corvallis, OR 97331, United States of America
| | - Bo Sun
- Department of Physics, Oregon State University, Corvallis, OR 97331, United States of America
| |
Collapse
|
44
|
Ebrahim NAA, Soliman SMA, Othman MO, Tahoun NS. Molecular mechanisms and clinical significance of perineural invasion in malignancies: the pivotal role of tumor-associated Schwann cells in cancer progression and metastasis. Med Oncol 2025; 42:171. [DOI: ebrahim, n.a.a., soliman, s.m.a., othman, m.o.et al.molecular mechanisms and clinical significance of perineural invasion in malignancies: the pivotal role of tumor-associated schwann cells in cancer progression and metastasis.med oncol 42, 171 (2025).https:/doi.org/10.1007/s12032-025-02729-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
|
45
|
Ebrahim NAA, Soliman SMA, Othman MO, Tahoun NS. Molecular mechanisms and clinical significance of perineural invasion in malignancies: the pivotal role of tumor-associated Schwann cells in cancer progression and metastasis. Med Oncol 2025; 42:171. [PMID: 40259163 DOI: 10.1007/s12032-025-02729-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: 02/06/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
Abstract
Perineural invasion (PNI) is a pathological process wherein cancer cells invade and spread along peripheral nerves, contributing to tumor aggressiveness and poor clinical outcomes, including increased recurrence, metastasis, and reduced survival. Tumor-associated Schwann cells (SCs) play a pivotal role in facilitating PNI by promoting epithelial-mesenchymal transition (EMT), extracellular matrix (ECM) remodeling, and immune modulation. These cells actively support tumor progression through neurotrophin, cytokine, chemokine, and neurotransmitter signaling, enhancing cancer cell migration along neural pathways. Recent advances in imaging techniques, single-cell transcriptomics, and molecular profiling have provided deeper insights into the tumor microenvironment's role in PNI. Emerging therapeutic strategies targeting neurotrophin-mediated signaling and SC-tumor interactions have shown promise in preclinical models. However, significant research gaps remain, particularly in understanding the heterogeneity of SCs and their molecular subtypes in PNI across different malignancies. This review highlights the clinical significance, molecular mechanisms, and potential therapeutic targets associated with PNI. A comprehensive understanding of tumor-SC interactions is essential for developing targeted interventions to mitigate PNI-driven malignancies. Future research should focus on integrating multi-omics approaches and novel therapeutics to improve early detection and treatment, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Noura A A Ebrahim
- Oncologic Pathology Department, National Cancer Institute (NCI) - Cairo University, Cairo, Egypt.
| | | | - Moamen O Othman
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Neveen S Tahoun
- Oncologic Pathology Department, National Cancer Institute (NCI) - Cairo University, Cairo, Egypt
| |
Collapse
|
46
|
Ebrahim NAA, Soliman SMA, Othman MO, Tahoun NS. Molecular mechanisms and clinical significance of perineural invasion in malignancies: the pivotal role of tumor-associated Schwann cells in cancer progression and metastasis. Med Oncol 2025; 42:171. [DOI: https:/doi.org/10.1007/s12032-025-02729-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
|
47
|
Mehta G, Bregenzer M, Mehta P, Burkhard K. Cracking the Code: Predicting Tumor Microenvironment Enabled Chemoresistance with Machine Learning in the Human Tumoroid Models. RESEARCH SQUARE 2025:rs.3.rs-5159414. [PMID: 40313776 PMCID: PMC12045351 DOI: 10.21203/rs.3.rs-5159414/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
High-grade serous tubo-ovarian cancer (HGSC) is marked by substantial inter- and intra-tumor heterogeneity. The tumor microenvironments (TME) of HGSC show pronounced variability in cellular make-up across metastatic sites, which is linked to poorer patient outcomes. The influence of cellular composition on therapy sensitivity, including chemotherapy and targeted treatments, has not been thoroughly investigated. In this study, we examined the premise that the variations in cellular composition can forecast drug efficacy. Using a high-throughput 3D in vitro tumoroid model, we assessed the drug responses of twenty-three distinct cellular configurations to an assortment of five therapeutic agents, including carboplatin and paclitaxel. By amalgamating our experimental findings with random forest machine learning algorithms, we assessed the influence of TME cellular composition on treatment reactions. Our findings reveal notable disparities in drug responses correlated with tumoroid composition, underscoring the significance of cellular diversity within the TME as a predictor of therapeutic outcomes. However, our work also emphasizes the complex nature of cell composition's influence on drug response. This research establishes a foundation for employing human tumoroids with varied cellular composition as a method to delve into the roles of stromal, immune, and other TME cell types in enhancing cancer cell susceptibility to various treatments. Additionally, these tumoroids can serve as a platform to explore pivotal cellular interactions within the TME that contribute to chemoresistance and cancer recurrence.
Collapse
|
48
|
Gao L, Jiang F. Streptococcus pneumoniae infection is associated with Matrix Metalloproteinase-9 in Lung Cancer Progression and Brain Metastases. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.15.25325903. [PMID: 40321254 PMCID: PMC12047908 DOI: 10.1101/2025.04.15.25325903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Matrix metalloproteinases (MMPs) are critical mediators of extracellular matrix remodeling, playing a pivotal role in the progression and metastasis of lung cancer. Emerging evidence indicates that bacterial pathogens, such as Streptococcus pneumoniae (SP), can modulate MMP activity and contribute to tumor progression. This study quantified SP abundance in lung cancer tissues and investigated its relationship with MMP-9 and MMP-2 expression, as well as its impact on clinical outcomes. SP DNA levels were assessed using droplet digital PCR, and MMP-9 and MMP-2 protein expression were evaluated by immunohistochemistry in 120 lung cancer samples. Elevated SP abundance was significantly associated with increased MMP-9 expression, advanced lung cancer stages, greater brain metastases burden, and reduced overall survival (P < 0.05). However, SP abundance showed no correlation with MMP-2 expression. These findings highlight a direct link between SP infection and lung cancer progression through MMP-9-mediated extracellular matrix degradation and metastatic spread. Targeting the SP-MMP-9 axis may represent a novel therapeutic approach to mitigate metastasis and improve patient outcomes in lung cancer.
Collapse
Affiliation(s)
- Lu Gao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Feng Jiang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
- BIOTARGET DX LLC, 7193 Somerton Ct, Hanover, Maryland 21076, USA
| |
Collapse
|
49
|
Chen W, Cheng Q, Li N, Gu K, Zhao H, Na H. The role of glycan-lectin interactions in the tumor microenvironment: immunosuppression regulators of colorectal cancer. Am J Cancer Res 2025; 15:1347-1383. [PMID: 40371166 PMCID: PMC12070101 DOI: 10.62347/wbjl4045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/17/2025] [Indexed: 05/16/2025] Open
Abstract
Colorectal cancer (CRC) is a common malignant tumour and a serious global health issue. Glycosylation, a type of posttranslational modification, has been extensively studied in relation to cancer growth and metastasis. Aberrant glycosylation alters how the immune system in the microenvironment perceives the tumour and drives immune suppression through glycan-binding receptors. Interestingly, specific glycan signatures can be regarded as a new pattern of immune checkpoints. Lectins are a group of proteins that exhibit high affinity for glycosylation structures. Lectins and their ligands are found on endothelial cells (ECs), immune cells and tumour cells and play important roles in the tumour microenvironment (TME). In CRC, glycan-lectin interactions can accelerate immune evasion promoting the differentiation of tumour-associated M2 macrophages, altering T cell, dendritic cell (DC), natural killer (NK) cell, and regulatory T (Treg) cell activity to modify the functions of antigen-presenting cells functions. Here, we review our current knowledge on how glycan-lectin interactions affect immune-suppressive circuits in the TME and discuss their roles in the development of more effective immunotherapies for CRC.
Collapse
Affiliation(s)
- Wenbin Chen
- Department of General Surgery, The People’s Hospital of China Medical University and The People’s Hospital of Liaoning ProvinceShenyang 110016, Liaoning, China
| | - Quanzhi Cheng
- Department of Laboratory Medicine, The People’s Hospital of China Medical University and The People’s Hospital of Liaoning ProvinceShenyang 110016, Liaoning, China
| | - Na Li
- Department of Laboratory Medicine, The People’s Hospital of China Medical University and The People’s Hospital of Liaoning ProvinceShenyang 110016, Liaoning, China
| | - Kaiming Gu
- Department of Laboratory Medicine, The People’s Hospital of China Medical University and The People’s Hospital of Liaoning ProvinceShenyang 110016, Liaoning, China
| | - Hongmei Zhao
- Department of Infection Management, The People’s Hospital of China Medical University and The People’s Hospital of Liaoning ProvinceShenyang 110016, Liaoning, China
| | - Heya Na
- Department of Laboratory Medicine, The People’s Hospital of China Medical University and The People’s Hospital of Liaoning ProvinceShenyang 110016, Liaoning, China
| |
Collapse
|
50
|
George S, Saju H, Jaikumar T, Raj R, Nisarga R, Sontakke S, Sangshetti J, Paul MK, Arote RB. Deciphering a crosstalk between biological cues and multifunctional nanocarriers in lung cancer therapy. Int J Pharm 2025; 674:125395. [PMID: 40064384 DOI: 10.1016/j.ijpharm.2025.125395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/08/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025]
Abstract
In recent years, the utilization of nanocarriers has significantly broadened across a diverse spectrum of biomedical applications. However, the clinical translation of these tiny carriers is limited and encounters hurdles, particularly in the intricate landscape of the tumor microenvironment. Lung cancer poses unique hurdles for nanocarrier design. Multiple physiological barriers hinder the efficient drug delivery to the lungs, such as the complex anatomy of the lung, the presence of mucus, immune responses, and rapid clearance mechanisms. Overcoming these obstacles necessitates a targeted approach that minimizes off-target effects while effectively penetrating nanoparticles/cargo into specific lung tissues or cells. Furthermore, understanding the cellular uptake mechanisms of these nano carriers is also essential. This knowledge aids in developing nanocarriers that efficiently enter cells and transfer their payload for the most effective therapeutic outcome. Hence, a thorough understanding of biological cues becomes crucial in designing multifunctional nanocarriers tailored for treating lung cancer. This review explores the essential biological cues critical for developing a flexible nanocarrier specifically intended to treat lung cancer. Additionally, it discusses advancements in nanotheranostics in lung cancer.
Collapse
Affiliation(s)
- Sharon George
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Hendry Saju
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Tharun Jaikumar
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Reshma Raj
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - R Nisarga
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Samruddhi Sontakke
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Jaiprakash Sangshetti
- Y. B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Rauza Baugh, Aurangabad 431001, India
| | - Manash K Paul
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India; Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), 90095 CA, USA.
| | - Rohidas B Arote
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India; Dental Research Institute, School of Dentistry, Seoul National University, Gwanak-ku, Seoul 08826, Republic of Korea.
| |
Collapse
|