1
|
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
|
2
|
Liang A, Tao T, Chen J, Yang Y, Zhou X, Zhu X, Yu G. Immunocompetent tumor-on-a-chip: A translational tool for drug screening and cancer therapy. Crit Rev Oncol Hematol 2025; 210:104716. [PMID: 40194716 DOI: 10.1016/j.critrevonc.2025.104716] [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: 02/18/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Tumor is one of the major diseases endangering human health while establishing an efficient in vitro tumor microenvironment (TME) model, which is an effective way to reveal the nature of the tumor and develop therapeutic methods. In recent years, due to the continuous development of lab-on-a-chip technology and tumor biology, various tumor-on-a-chip models applied to oncology research have emerged. Among them, the Immunotherapy-on-a-chip (ITOC) platform stands out with its ability to reflect immunological behavior in the TME. It is a class of in vitro tumor-on-a-chip with immune activity, which has good performance and the ability to reproduce TME. It can highly simulate the complex pathophysiological characteristics of tumors and be used to study various features related to tumor biological behavior. Currently, many advantageous functions and application values of ITOC platforms have been discovered and applied to tumor drug screening and development, tumor immunotherapy, and personalized therapy. In conclusion, the tumor-on-a-chip platform is a highly promising model for medical oncology research. In this review, the background of the ITOC platform, key factors for constructing an ideal ITOC platform, and the specific applications of ITOC platforms in tumor research and treatment are introduced.
Collapse
Affiliation(s)
- Anqi Liang
- Department of Cardiothoracic Surgery, Jiangyin People's Hospital Affiliated to Nantong University, Jiangyin, China; The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Jiahui Chen
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Yucong Yang
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Xiao Zhu
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China.
| | - Guiping Yu
- Department of Cardiothoracic Surgery, Jiangyin People's Hospital Affiliated to Nantong University, Jiangyin, China.
| |
Collapse
|
3
|
Sun L, Liu Y, Sun Q, Wang G, Du B, Liu B, Gao T, Zhao P, Yang Y, Rong R. Polysaccharides from traditional Chinese medicine and their nano-formulated delivery systems for cancer immunotherapy. Carbohydr Polym 2025; 357:123416. [PMID: 40158963 DOI: 10.1016/j.carbpol.2025.123416] [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: 11/25/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 04/02/2025]
Abstract
Cancer immunotherapy has evolved into a new generation strategy in the field of anti-tumor treatment. Polysaccharides derived from Traditional Chinese Medicine (TCM) are gaining recognition as powerful immunomodulators in cancer therapy, noted for their multi-target and multi-pathway actions. Owing to their beneficial properties such as water solubility, biocompatibility, and chemical structure modifiability, TCM polysaccharides can also serve as carriers for hydrophobic drugs in the development of innovative drug delivery systems, enhancing synergistic antitumor effects. In this article, we summarize the diverse mechanisms of immunoregulation by TCM polysaccharides in tumor therapy. The applications of these polysaccharides as both active ingredients and drug carriers within nanodelivery systems for cancer immunotherapy are also introduced. Additionally, extensive research on TCM polysaccharides in clinical settings has been collected. Furthermore, discussions are presented on the development prospects and challenges faced by these polysaccharides in the field of tumor immunotherapy. Our goal is to improve researchers' comprehension of TCM polysaccharides in cancer immunotherapy, providing promising strategies to optimize cancer treatment and benefit diverse patient populations.
Collapse
Affiliation(s)
- Linlin Sun
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yuting Liu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Qihui Sun
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Guimei Wang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Baoxiang Du
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Bodong Liu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Tian Gao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Pan Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yong Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China; Collaborative Innovation Center for Antiviral Traditional Chinese Medicine in Shandong Province, Jinan 250355, PR China; Shandong Antiviral Engineering Research Center of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Rong Rong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| |
Collapse
|
4
|
Fang Y, Tan C, Zheng Z, Yang J, Tang J, Guo R, Silli EK, Chen Z, Chen J, Ge R, Liu Y, Wen X, Liang J, Zhu Y, Jin Y, Li Q, Wang Y. The function of microRNA related to cancer-associated fibroblasts in pancreatic ductal adenocarcinoma. Biochem Pharmacol 2025; 236:116849. [PMID: 40056941 DOI: 10.1016/j.bcp.2025.116849] [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: 11/11/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignant tumor characterized by a poor prognosis. A prominent feature of PDAC is the rich and dense stroma present in the tumor microenvironment (TME), which significantly hinders drug penetration. Cancer-associated fibroblasts (CAFs), activated fibroblasts originating from various cell sources, including pancreatic stellate cells (PSCs) and mesenchymal stem cells (MSCs), play a critical role in PDAC progression and TME formation. MicroRNAs (miRNAs) are small, single-stranded non-coding RNA molecules that are frequently involved in tumorigenesis and progression, exhibiting either oncolytic or oncogenic activity. Increasing evidence suggests that aberrant expression of miRNAs can mediate interactions between cancer cells and CAFs, thereby providing novel therapeutic targets for PDAC treatment. In this review, we will focus on the potential roles of miRNAs that target CAFs or CAFs-derived exosomes in PDAC progression, highlighting the feasibility of therapeutic strategies aimed at restoring aberrantly expressed miRNAs associated with CAFs, offering new pathways for the clinical management of PDAC.
Collapse
Affiliation(s)
- Yaohui Fang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Chunlu Tan
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenjiang Zheng
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianchen Yang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jiali Tang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruizhe Guo
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Epiphane K Silli
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Zhe Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jia Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruyu Ge
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yuquan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xiuqi Wen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jingdan Liang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yunfei Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yutong Jin
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Qian Li
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ying Wang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| |
Collapse
|
5
|
Xu Y, Li X, Cui M, Pan T, Zheng S, Shang Z, Yin D, Xu Y, Yang Z, Wang J, Song X. Fufang-Biejia-Ruangan tablet targeting both cancer-associated fibroblasts and tumor cells by HIPPO-PI3K/AKT cascades in intrahepatic cholangiocarcinoma treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156690. [PMID: 40215817 DOI: 10.1016/j.phymed.2025.156690] [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: 01/03/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (ICC) ranks second among primary liver cancers in terms of prevalence, characterized by poor prognosis and scarce therapeutic interventions. Fufang-Biejia-Ruangan tablet (BJRG), a traditional Chinese herbal remedy, has been widely used for liver diseases. However, its therapeutic efficacy and underlying mechanisms in ICC remain poorly understood. AIM OF THE STUDY This study aims to systematically investigate the anti-ICC effects of BJRG, focusing on tumor progression and microenvironment modulation, through experimental and transcriptomic analyses. METHODS The chemical composition of BJRG was analyzed employing ultra-performance liquid chromatography-mass spectrometry (UPLC-MS). In vitro assays were performed with QBC939 and LX-2 cell lines. Two primary ICC models (AKT/YAP and sgP53/KRAS) were established via hydrodynamic tail-vein injection of corresponding plasmids. A co-culture system for subcutaneous tumor formation was developed using cancer-associated fibroblasts (CAFs) derived from the AKT/YAP model and primary tumor cells derived from the sgP53/KRAS model. RESULTS UPLC-MS analysis identified 1091 chemical components, primarily terpenoids, sugars, glycosides, and phenylpropanoids. The therapeutic efficacy of BJRG was evaluated for the treatment of ICC. BJRG treatment slowed down the growth of both human ICC cell lines and AKT/YAP ICC mouse model. Mechanistically, BJRG inhibited HIPPO-PI3K/AKT signaling pathway in ICC tumor cells. Importantly, BJRG significantly inhibited the growth of CAFs via HIPPO-PI3K/AKT cascades. Of note, co-culture CAFs with ICC cell lines substantially sensitized the efficacy of BJRG in sgP53/KRAS syngeneic tumor model. Furthermore, BJRG therapy not only affected CAFs but also induced alterations in vascular structures and hypoxic conditions within lesions in the AKT/YAP model. This intervention promoted the infiltration of T lymphocytes and macrophages into the tumor microenvironment, which may further augment the anti-proliferative effects of BJRG by enhancing the immune response within ICC tumor tissues. CONCLUSION Our research demonstrates BJRG's anti-ICC efficacy via diverse pathways, including the suppression of tumor cell proliferation, regulation of CAFs activity, and promotion of immune cell infiltration. These findings underscore BJRG as a promising therapeutic candidate for ICC, offering novel mechanistic insights and highlighting its potential for clinical translation.
Collapse
Affiliation(s)
- Yanyu Xu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350122, China; Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Xiang Li
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Miao Cui
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Tao Pan
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Shuwen Zheng
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350122, China; Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Zimeng Shang
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; Center of Integrative Medicine, Beijing Ditan Hospital, Affiliated to Capital Medical University, Beijing 100015, China
| | - Donghao Yin
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; Beijing You'an Hospital, Affiliated to Capital Medical University, Beijing, 100069, China
| | - Yang Xu
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Zhiyun Yang
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; Center of Integrative Medicine, Beijing Ditan Hospital, Affiliated to Capital Medical University, Beijing 100015, China
| | - Jiabo Wang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350122, China; Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China.
| | - Xinhua Song
- Laboratory for Clinical Medicine, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China; School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
6
|
Oikawa Y, Umakoshi M, Suzuki K, Kudo-Asabe Y, Miyabe K, Koyama K, Yoshida M, Tanaka M, Nanjo H, Fukuda M, Yamada T, Goto A. Prognostic significance of cancer-associated fibroblasts and tumor-associated macrophages in the tongue squamous cell carcinoma and their correlation with tumor budding. Oral Oncol 2025; 165:107295. [PMID: 40327897 DOI: 10.1016/j.oraloncology.2025.107295] [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: 11/18/2024] [Revised: 03/26/2025] [Accepted: 04/05/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is the most prevalent malignancy of the oral cavity and is characterized by a high propensity for invasion and a poor prognosis. Recent studies have highlighted the critical roles of cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs) in tumor progression, particularly within the tumor microenvironment (TME). OBJECTIVE This study aimed to investigate the correlation between CAFs, TAMs, and tumor budding in tongue squamous cell carcinoma (TSCC), and evaluate their impact on prognostic factors. METHODS A total of 88 cases of surgically resected TSCC were analyzed. Immunohistochemical staining was performed using markers of CAFs (fibroblast activation protein, FAP) and TAMs (CD163). The correlation between CAF and TAM scores, tumor budding, and various clinicopathological factors was assessed. TAM scores were evaluated for the number of TAMs in the intratumoral areas (TAM-t) and the invasive front (TAM-fr). CAF scores were evaluated for cancer cells in the intratumoral area (cCAF-t), stromal cells in the intratumoral area (sCAF-t), stromal cells in the invasive front (sCAF-fr), and the infiltration pattern of CAF (IPC). RESULTS The IPC score was significantly associated with the tumor budding scores (p < 0.001) and poor DFS (p < 0.01). In the multivariate analysis, cCAF-t, sCAF-t, and IPC scores emerged as independent prognostic factors (p < 0.05) for early-stage TSCC. CAFs may play a pivotal role in tumor invasion. CONCLUSION These findings indicate that CAFs significantly influence the invasive characteristics of TSCC and are correlated with tumor budding and a poor prognosis. These results underscore the potential of targeting CAFs as a therapeutic strategy for improving OSCC outcome.
Collapse
Affiliation(s)
- Yuki Oikawa
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan; Department of Dentistry and Oral Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Michinobu Umakoshi
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan; Department of Pathology, Akita City Hospital, Akita, Japan.
| | - Kenichiro Suzuki
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan; Department of Dentistry and Oral Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Yukitsugu Kudo-Asabe
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Ken Miyabe
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Kei Koyama
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Makoto Yoshida
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Masamitsu Tanaka
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| | - Hiroshi Nanjo
- Department of Pathology, Akita University Hospital, Akita, Japan
| | - Masayuki Fukuda
- Department of Dentistry and Oral Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Takechiyo Yamada
- Department of Otorhinolaryngology, Head and Neck Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Akiteru Goto
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
7
|
Cai G, Rodgers NC, Liu AP. Unjamming Transition as a Paradigm for Biomechanical Control of Cancer Metastasis. Cytoskeleton (Hoboken) 2025; 82:388-403. [PMID: 39633605 PMCID: PMC12137693 DOI: 10.1002/cm.21963] [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: 08/14/2024] [Revised: 10/27/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
Tumor metastasis is a complex phenomenon that poses significant challenges to current cancer therapeutics. While the biochemical signaling involved in promoting motile phenotypes is well understood, the role of biomechanical interactions has recently begun to be incorporated into models of tumor cell migration. Specifically, we propose the unjamming transition, adapted from physical paradigms describing the behavior of granular materials, to better discern the transition toward an invasive phenotype. In this review, we introduce the jamming transition broadly and narrow our discussion to the different modes of 3D tumor cell migration that arise. Then we discuss the mechanical interactions between tumor cells and their neighbors, along with the interactions between tumor cells and the surrounding extracellular matrix. We center our discussion on the interactions that induce a motile state or unjamming transition in these contexts. By considering the interplay between biochemical and biomechanical signaling in tumor cell migration, we can advance our understanding of biomechanical control in cancer metastasis.
Collapse
Affiliation(s)
- Grace Cai
- Applied Physics ProgramUniversity of MichiganAnn ArborMichiganUSA
| | - Nicole C. Rodgers
- Department of Mechanical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Allen P. Liu
- Applied Physics ProgramUniversity of MichiganAnn ArborMichiganUSA
- Department of Mechanical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of BiophysicsUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
8
|
Kazmi A, Gill R, Restrepo P, Ji AL. The spatial and single-cell landscape of skin: Charting the multiscale regulation of skin immune function. Semin Immunol 2025; 78:101958. [PMID: 40267702 PMCID: PMC12146056 DOI: 10.1016/j.smim.2025.101958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
Immune regulation is a key function of the skin, a barrier tissue that exhibits spatial compartmentalization of innate and adaptive immune cells. Recent advances in single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST) have facilitated systems-based investigations into the molecular and cellular features of skin immunity at single-cell resolution, identifying cell types that maintain homeostasis in a coordinated manner, and those that exhibit dysfunctional cell-cell interactions in disease. Here, we review how technological innovation is uncovering the multiple scales of heterogeneity in the immune landscape of the skin. The microanatomic scale encompasses the skin's diverse cellular components and multicellular spatial organization, which govern the functional cell interactions and behaviors necessary to protect the host. On the macroanatomic scale, understanding heterogeneity in cutaneous tissue architecture across anatomical sites promises to unearth additional functional immune variation and resulting disease consequences. We focus on how single-cell and spatial dissection of the immune system in experimental models and in humans has led to a deeper understanding of how each cell type in the skin contributes to overall immune function in a context-dependent manner. Finally, we highlight translational opportunities for adopting these technologies, and insights gleaned from them, into the clinic.
Collapse
Affiliation(s)
- Abiha Kazmi
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raman Gill
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paula Restrepo
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew L Ji
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
9
|
Vera RE, Fernadez-Barrena MG, Falero JM, Kwon JY, Garza RA, Sigafoos AN, Ross MD, Toruner MD, Toruner M, Tolosa EJ, Almada LL, Huang H, Brekken RA, Fernandez-Zapico ME. Paracrine regulation of pancreatic cancer cell response to chemotherapy by GLI2-Collagen I signaling. J Biol Chem 2025:110311. [PMID: 40449600 DOI: 10.1016/j.jbc.2025.110311] [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: 12/12/2024] [Revised: 05/08/2025] [Accepted: 05/25/2025] [Indexed: 06/03/2025] Open
Abstract
Despite the well described role of non-cellular components of the tumor microenvironment (TME) in regulating tumor growth, the molecular events dictating expression and biological functions of key components of the TME remain elusive. Here, using pancreatic cancer (PC) models, we describe a novel mechanism through which the zinc finger transcription factor GLI2 in cancer associated fibroblasts (CAFs) induces expression of COL1A1, which is a major component of Type I Collagen, the most abundant collagen variant in the tumor milieu. Bulk and single nuclei RNA-Seq showed that GLI2 expression in CAF strongly correlates with COL1A1 expression levels, fibrosis, and CAF activation. ChIP-qPCR and expression studies of the PC matrisome identified COL1A1 as the direct target of GLI2 in CAFs. We also provide evidence that GLI2 is an effector that mediates COL1A1 induction by transforming growth factor β1 (TGFβ1). RNA-Seq analysis of PC cells treated with Type I Collagen revealed enrichment of chemotherapeutic gene expression profiles, which includes irinotecan resistance signature. Viability studies confirmed that Type I Collagen promotes irinotecan resistance in PC cells. Altogether, our results uncover a novel role for the TGFβ1-GLI2 axis within CAFs to modulate Type I Collagen expression and promote chemoresistance in PC cells. Together, our findings help increase the understanding of the complex molecular network operating in the TME.
Collapse
Affiliation(s)
- Renzo E Vera
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN
| | | | - Jose M Falero
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN
| | - John Y Kwon
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN
| | - Roberto A Garza
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN
| | - Ashley N Sigafoos
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN
| | - Matthew D Ross
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN
| | - Merih Deniz Toruner
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN
| | - Murat Toruner
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN
| | - Ezequiel J Tolosa
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN
| | - Luciana L Almada
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN
| | - Huocong Huang
- Departments of Surgery and Immunology, Hamon Center for Therapeutic Oncology Research, UT Southwestern, Dallas, TX
| | - Rolf A Brekken
- Departments of Surgery and Pharmacology, Hamon Center for Therapeutic Oncology Research, UT Southwestern, Dallas, TX
| | | |
Collapse
|
10
|
Lee JJ, Ng KY, Bakhtiar A. Extracellular matrix: unlocking new avenues in cancer treatment. Biomark Res 2025; 13:78. [PMID: 40426238 PMCID: PMC12117852 DOI: 10.1186/s40364-025-00757-3] [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: 10/14/2024] [Accepted: 03/05/2025] [Indexed: 05/29/2025] Open
Abstract
The extracellular matrix (ECM) plays a critical role in cancer progression by influencing tumor growth, invasion, and metastasis. This review explores the emerging therapeutic strategies that target the ECM as a novel approach in cancer treatment. By disrupting the structural and biochemical interactions within the tumor microenvironment, ECM-targeted therapies aim to inhibit cancer progression and overcome therapeutic resistance. We examine the current state of ECM research, focusing on key components such as collagen, laminin, fibronectin, periostin, and hyaluronic acid, and their roles in tumor biology. Additionally, we discuss the challenges associated with ECM-targeted therapies, including drug delivery, specificity, and potential side effects, while highlighting recent advancements and future directions. This review underscores the potential of ECM-focused strategies to enhance the efficacy of existing treatments and contribute to more effective cancer therapies.
Collapse
Affiliation(s)
- Jia Jing Lee
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Athirah Bakhtiar
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia.
| |
Collapse
|
11
|
Krönke T, Kopka K, Mamat C. Enhancing the radionuclide theranostic concept through the radiohybrid approach. RSC Med Chem 2025; 16:1856-1864. [PMID: 39618962 PMCID: PMC11606402 DOI: 10.1039/d4md00591k] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/25/2024] [Indexed: 06/01/2025] Open
Abstract
Radionuclide theranostics - a fast-growing emerging field in radiopharmaceutical sciences and nuclear medicine - offers a personalised and precised treatment approach by combining diagnosis with specific and selective targeted endoradiotherapy. This concept is based on the application of the same molecule, labelled with radionuclides possessing complementary imaging and therapeutic properties, respectively. In radionuclide theranostics, radionuclide pairs consisting of the same element, such as 61/64Cu/67Cu, 203Pb/212Pb or 123/124I/131I are of significant interest due to their identical chemical and pharmacological characteristics. However, such "true matched pairs" are seldom, necessitating the use of complementary radionuclides from different elements for diagnostics and endoradiotherapy with similar chemical characteristics, such as 99mTc/186/188Re, 68Ga/177Lu or 68Ga/225Ac. Corresponding combinations of such two radionuclides in one and the same radioconjugate is referred to as a "matched pair". Notably, the pharmacological behavior remains consistent across both diagnostic and therapeutic applications with "true matched pairs", which may differ for "matched pairs". As "true matched pairs" of theranostic radioisotopes are rare and that some relevant radionuclides do not fit with the diagnostic or therapeutic counterpart, the radionuclide theranostic concept can be expanded and improved by the introduction of the radiohybrid approach. Radiohybrid (rh) ligands represent a new class of radiopharmaceutical bearing two different positions for the introduction of a (radio)metal and (radio)halogen in one molecule, which can be then used for both therapeutic and diagnostic purposes. The following review will give an insight into recent developments of this approach.
Collapse
Affiliation(s)
- Tobias Krönke
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstraße 400 D-01328 Dresden Germany
- TU Dresden, Faculty of Chemistry and Food Chemistry D-01062 Dresden Germany
| | - Klaus Kopka
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstraße 400 D-01328 Dresden Germany
- TU Dresden, Faculty of Chemistry and Food Chemistry D-01062 Dresden Germany
- National Center for Tumor Diseases (NCT) Dresden, University Hospital Carl Gustav Carus Fetscherstraße 74 D-01307 Dresden Germany
- German Cancer Consortium (DKTK) Partner Site Dresden, Fetscherstraße 74 D-01307 Dresden Germany
| | - Constantin Mamat
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstraße 400 D-01328 Dresden Germany
- TU Dresden, Faculty of Chemistry and Food Chemistry D-01062 Dresden Germany
| |
Collapse
|
12
|
Wang S, Zheng Q, Chen L. Integration of Bulk and Single-Cell RNA Sequencing to Identify RNA Modifications-Related Prognostic Signature in Ovarian Cancer. Int J Gen Med 2025; 18:2629-2647. [PMID: 40417417 PMCID: PMC12103173 DOI: 10.2147/ijgm.s523878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025] Open
Abstract
Background Ovarian cancer (OC), a common fatal malignancy in women, has a poor prognosis. RNA modifications are associated with the development of OC. In this study, we aimed to identify and verify RNA modifications-related prognostic genes in OC by integrating bulk and single-cell RNA sequencing (scRNA-seq) data. Methods Transcriptome data came from public databases and RNA modifications-related genes (RMRGs) were obtained from literature. Candidate genes were identified by intersecting RMRGs with differentially expressed genes (DEGs) in OC patients. Prognostic genes were gained via machine learning techniques, particularly LASSO regression. A risk model was built to predict the prognosis. OC patients were divided into high-risk and low-risk groups according to risk score. Subsequent analyses covered enrichment analysis, immune microenvironment, mutation analysis, and chemotherapeutic drug sensitivity. In addition, scRNA-seq data was assessed for key cells and gene expression in them. Finally, RT-qPCR was applied to identify the expression of prognostic genes. Results LSM4, SNRPC, ZC3H13, LSM2, WTAP, DCP2, PUS7, and TUT1 were selected as prognostic genes. The risk model exhibited excellent predictive abilities. Seventeen pathways were enriched like calcium signaling pathway, 7 differential immune cells were identified like regulatory T cells and plasmacytoid dendritic cells, and TP53 had highest mutation rate. Half-maximal inhibitory concentrations (IC50) values of 47 drugs like paclitaxel differed between two risk groups. The prognostic genes were distributed mainly in fibroblast cells, epithelial cells and endothelial cells. During fibroblast cells differentiation, expression of prognostic genes fluctuated to varying degrees. The RT-qPCR demonstrated that the expression of LSM2, LSM4, PUS7, SNRPC, and TUT1 were upregulated in OC, while DCP2, WTAP, and ZC3H13 were downregulated. Conclusion We constructed an RNA modifications-related prognostic signature that can effectively predict clinical outcomes and therapeutic responses in patients with OC.
Collapse
Affiliation(s)
- Shaoyu Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Obstetrics and Gynecology, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
| | - Qiaomei Zheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Obstetrics and Gynecology, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
| | - Lihong Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of China
- Department of Obstetrics and Gynecology, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, People’s Republic of China
- Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People’s Republic of 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
|
Aggarwal A, Jana M, Singh A, Dam T, Maurya H, Pathak T, Orsulic S, Yang K, Chute D, Bishop JA, Faraji F, Thorstad WM, Koyfman S, Steward S, Shi Q, Sandulache V, Saba NF, Lewis JS, Corredor G, Madabhushi A. Artificial intelligence-based virtual staining platform for identifying tumor-associated macrophages from hematoxylin and eosin-stained images. Eur J Cancer 2025; 220:115390. [PMID: 40158294 PMCID: PMC12021545 DOI: 10.1016/j.ejca.2025.115390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Virtual staining is an artificial intelligence-based approach that transforms pathology images between stain types, such as hematoxylin and eosin (H&E) to immunohistochemistry (IHC), providing a tissue-preserving and efficient alternative to traditional IHC staining. However, existing methods for translating H&E to virtual IHC often fail to generate images of sufficient quality for accurately delineating cell nuclei and IHC+ regions. To address these limitations, we introduce VISTA, an artificial intelligence-based virtual staining platform designed to translate H&E into virtual IHC. METHODS We applied VISTA to identify M2-subtype tumor-associated macrophages (M2-TAMs) in H&E images from 968 patients with HPV+ oropharyngeal squamous cell carcinoma across six institutional cohorts. M2-TAMs are a critical component of the tumor microenvironment, and their increased presence has been linked to poor survival. Co-registered H&E and CD163 + IHC tissue microarrays were used to train (D1, N = 102) and test (D2, N = 50) the VISTA platform. M2-TAM density, defined as the ratio of M2-TAMs to total nuclei, was derived from VISTA-generated CD163 + IHC images and evaluated for prognostic significance in additional training (D3, N = 360) and testing (D4, N = 456) cohorts using biopsy or resection H&E whole slide images. RESULTS High M2-TAM density was associated with worse overall survival in D4 (p = 0.0152, Hazard Ratio=1.63 [1.1-2.42]). VISTA outperformed existing methods, generating higher-quality virtual CD163 + IHC images in D2, with a Structural Similarity Index of 0.72, a Peak Signal-to-Noise Ratio of 21.5, and a Fréchet Inception Distance of 41.4. Additionally, VISTA demonstrated superior performance in segmenting M2-TAMs in D2 (Dice=0.74). CONCLUSION These findings establish VISTA as a computational platform for generating virtual IHC and facilitating the discovery of novel biomarkers from H&E images.
Collapse
Affiliation(s)
- Arpit Aggarwal
- Department of Biomedical Engineering, Georgia Tech, GA, USA; Department of Biomedical Engineering, Emory University, GA, USA
| | - Mayukhmala Jana
- Department of Biomedical Engineering, Georgia Tech, GA, USA; Department of Biomedical Engineering, Emory University, GA, USA
| | - Amritpal Singh
- Department of Biomedical Engineering, Emory University, GA, USA
| | - Tanmoy Dam
- Department of Biomedical Engineering, Emory University, GA, USA
| | - Himanshu Maurya
- Department of Biomedical Engineering, Emory University, GA, USA
| | - Tilak Pathak
- Department of Biomedical Engineering, Emory University, GA, USA
| | | | - Kailin Yang
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA
| | - Deborah Chute
- Department of Anatomic Pathology, Cleveland Clinic, Cleveland, OH, USA
| | - Justin A Bishop
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Farhoud Faraji
- Department of Otolaryngology-Head and Neck Surgery, UC San Diego Health, La Jolla, CA, USA
| | - Wade M Thorstad
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MS, USA
| | - Shlomo Koyfman
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Scott Steward
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, GA, USA
| | - Qiuying Shi
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University School of Medicine, OR, USA
| | - Vlad Sandulache
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, TX, USA; ENT Section, Operative CareLine, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Nabil F Saba
- Department of Hematology and Medical Oncology, Emory University School of Medicine, GA, USA
| | - James S Lewis
- Department of Laboratory Medicine and Pathology, Mayo Clinic, AZ, USA
| | - Germán Corredor
- Department of Biomedical Engineering, Georgia Tech, GA, USA; Department of Biomedical Engineering, Emory University, GA, USA; Atlanta VA Medical Center, GA, USA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Georgia Tech, GA, USA; Department of Biomedical Engineering, Emory University, GA, USA; Atlanta VA Medical Center, GA, USA.
| |
Collapse
|
15
|
Aquino A, Franzese O. Reciprocal Modulation of Tumour and Immune Cell Motility: Uncovering Dynamic Interplays and Therapeutic Approaches. Cancers (Basel) 2025; 17:1547. [PMID: 40361472 PMCID: PMC12072109 DOI: 10.3390/cancers17091547] [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: 03/31/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Dysregulated cell movement is a hallmark of cancer progression and metastasis, the leading cause of cancer-related mortality. The metastatic cascade involves tumour cell migration, invasion, intravasation, dissemination, and colonisation of distant organs. These processes are influenced by reciprocal interactions between cancer cells and the tumour microenvironment (TME), including immune cells, stromal components, and extracellular matrix proteins. The epithelial-mesenchymal transition (EMT) plays a crucial role in providing cancer cells with invasive and stem-like properties, promoting dissemination and resistance to apoptosis. Conversely, the mesenchymal-epithelial transition (MET) facilitates metastatic colonisation and tumour re-initiation. Immune cells within the TME contribute to either anti-tumour response or immune evasion. These cells secrete cytokines, chemokines, and growth factors that shape the immune landscape and influence responses to immunotherapy. Notably, immune checkpoint blockade (ICB) has transformed cancer treatment, yet its efficacy is often dictated by the immune composition of the tumour site. Elucidating the molecular cross-talk between immune and cancer cells, identifying predictive biomarkers for ICB response, and developing strategies to convert cold tumours into immune-active environments is critical to overcoming resistance to immunotherapy and improving patient survival.
Collapse
Affiliation(s)
| | - Ornella Franzese
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| |
Collapse
|
16
|
Demirkol Canlı S, Güner G, Işık A, Sosic-Jurjevi B, Rom AD, Seza EG, Dizdar Ö, Dragicevic S, Nikolic A, Akyol A, Banerjee S. AKR1B1 Expression in the Colorectal Tumor Microenvironment Contributes Towards Its Prognostic Significance. Cancer Med 2025; 14:e70974. [PMID: 40396420 DOI: 10.1002/cam4.70974] [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: 01/23/2025] [Revised: 05/06/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND AKR1B1, a member of the aldo-keto reductase enzyme family involved in the polyol pathway of aldehyde metabolism, is aberrantly expressed in colorectal cancer (CRC). Our previous studies demonstrated that AKR1B1 knockdown reduced the motility and proliferation of CRC cell lines, and its elevated expression was correlated with increased mesenchymal marker expression, inflammation, and poor prognosis in CRC patient cohorts. However, whether stromal cells also express AKR1B1 and whether stromal expression can affect clinical outcomes has not been examined. OBJECTIVES To evaluate the expression of AKR1B1 within the tumor microenvironment (TME) of CRC, with a paticular focus on stromal cells, and to assess its association with clinical outcomes. METHODS We assessed AKR1B1 expression in colorectal tumors utilizing publicly available transcriptomic data from CRC tumors. Single-cell RNA-sequencing data from CRC samples were analyzed to determine cell type-specific expression. Immunohistochemistry based assessment of AKR1B1 expression was performed in Turkish and Serbian cohorts. Co-localization of AKR1B1 and CD163 (M2 macrophage marker) was evaluated by immunoflourescence. RESULTS AKR1B1 was expressed in both the epithelial and stromal components of colorectal tumors, with higher expression observed in the stroma. Single-cell transcriptomic analysis revealed AKR1B1 expression in myeloid cells, T and NK cells, B cells, dendritic cells, fibroblasts, and epithelial cells. Notably, AKR1B1-expressing macrophages were predominantly of the M2 phenotype, and AKR1B1 expression and M2 marker expression showed strong positive correlation in bulk transcriptomic data. Immunofluorescence confirmed the colocalization of CD163 and AKR1B1 in stromal macrophages. Moreover, immunohistochemical analysis of AKR1B1 expression in tumor stroma from a cohort of Turkish patients revealed that its expression was associated with favorable overall survival, particularly in tumors with higher stromal infiltration. CONCLUSIONS Overall, our findings underscore the significant influence of the TME composition on the relationship between AKR1B1 expression and clinical outcomes.
Collapse
Affiliation(s)
- Seçil Demirkol Canlı
- Division of Tumor Pathology, Department of Clinical Oncology, Cancer Institute, Hacettepe University, Ankara, Turkiye
| | - Güneş Güner
- Department of Pathology, Hacettepe University Faculty of Medicine, Ankara, Turkiye
| | - Aynur Işık
- Transgenic Animal Technologies Research and Application Center, Hacettepe University, Ankara, Turkiye
| | - Branka Sosic-Jurjevi
- Department of Cytology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Djikic Rom
- Department of Pathology, Pathohistology and Medical Cytology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Esin Gülce Seza
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkiye
| | - Ömer Dizdar
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkiye
| | - Sandra Dragicevic
- Gene Regulation in Cancer Group, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Nikolic
- Gene Regulation in Cancer Group, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Aytekin Akyol
- Department of Pathology, Hacettepe University Faculty of Medicine, Ankara, Turkiye
- Transgenic Animal Technologies Research and Application Center, Hacettepe University, Ankara, Turkiye
| | - Sreeparna Banerjee
- Department of Biological Sciences, Orta Dogu Teknik Universitesi, Ankara, Turkiye
| |
Collapse
|
17
|
Guo R, Wang P. Tumor-derived extracellular vesicles: Hijacking T cell function through exhaustion. Pathol Res Pract 2025; 269:155948. [PMID: 40168777 DOI: 10.1016/j.prp.2025.155948] [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: 01/20/2025] [Revised: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Extracellular vesicles (EVs) play a vital role in intercellular communication within the tumor microenvironment (TME). These vesicles, secreted by tumor cells, contain proteins, lipids, and nucleic acids that significantly influence immune responses, particularly impacting T-cell function. In cancer, T cell dysfunction and exhaustion-marked by reduced proliferation, diminished cytokine production, and impaired cytotoxic activity-are key barriers to effective immune responses. Tumor-derived extracellular vesicles (TEVs) contribute to this dysfunction by carrying immunosuppressive molecules, such as transforming growth factor-beta (TGF-β) and various microRNAs (miRNAs). These TEV-mediated mechanisms promote T cell exhaustion and foster a broader immunosuppressive environment, enabling tumor progression and immune evasion. Furthermore, TEVs have been implicated in resistance to cancer immunotherapies, including immune checkpoint inhibitors and T cell therapies. Understanding the molecular pathways and cargoes within TEVs that drive T cell dysfunction is crucial for developing novel therapeutic strategies aimed at reinvigorating exhausted T cells, enhancing anti-tumor immunity, and improving cancer treatment outcomes.
Collapse
Affiliation(s)
- RuiJuan Guo
- Department of Oncology, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, Shandong 264003, China
| | - Ping Wang
- Department of Oncology, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, Shandong 264003, China.
| |
Collapse
|
18
|
Torregrossa M, Davies L, Hans-Günther M, Simon JC, Franz S, Rinkevich Y. Effects of embryonic origin, tissue cues and pathological signals on fibroblast diversity in humans. Nat Cell Biol 2025; 27:720-735. [PMID: 40263573 DOI: 10.1038/s41556-025-01638-5] [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: 01/22/2024] [Accepted: 02/18/2025] [Indexed: 04/24/2025]
Abstract
Fibroblasts, once perceived as a uniform cell type, are now recognized as a mosaic of distinct populations with specialized roles in tissue homeostasis and pathology. Here we provide a global overview of the expanding compendium of fibroblast cell types and states, their diverse lineage origins and multifaceted functions across various human organs. By integrating insights from developmental biology, lineage tracing and single-cell technologies, we highlight the complex nature of fibroblasts. We delve into their origination from embryonic mesenchyme and tissue-resident populations, elucidating lineage-specific behaviours in response to physiological cues. Furthermore, we highlight the pivotal role of fibroblasts in orchestrating tissue repair, connective tissue remodelling and immune modulation across diverse pathologies. This knowledge is essential to develop novel fibroblast-targeted therapies to restore steady-state fibroblast function and advance regenerative medicine strategies across multiple diseases.
Collapse
Affiliation(s)
- Marta Torregrossa
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany
| | - Lindsay Davies
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Machens Hans-Günther
- Department for Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Jan C Simon
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany
| | - Sandra Franz
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany.
| | - Yuval Rinkevich
- Chinese Institutes for Medical Research, Beijing, China.
- Capital Medical University, Beijing, China.
| |
Collapse
|
19
|
Maubach G, Kanthasamy AK, Gogia S, Naumann M. The enigma of maladaptation in gastric pathophysiology. Trends Cancer 2025; 11:448-461. [PMID: 39984410 DOI: 10.1016/j.trecan.2025.01.014] [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: 11/14/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/23/2025]
Abstract
Despite a decline in global incidence, gastric cancer (GC) remains a major health concern. The development of GC is a sequential, multistage maladaptive process involving numerous different factors. Understanding the complexity of GC development is crucial for early detection, effective treatment, and, ultimately, prevention. In this respect, identifying the impact of risk factors contributing to the emergence or progression of GC, such as Helicobacter pylori infection, host and bacterial genetics, alcohol consumption, smoking, and preserved foods, will aid in combatting this disease. In this review, we focus on recent developments in understanding the role of the microbiome, dysfunctional molecular pathways, and immune evasion in gastric pathophysiology. We also highlight challenges and advances in treatment of GC.
Collapse
Affiliation(s)
- Gunter Maubach
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Arun K Kanthasamy
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Sandro Gogia
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany.
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Yin X, Zhao X, Shen Y, Xie W, He C, Guo J, Li Z, Xuan F, Zeng S, Zeng X, Fang C. Nanoparticle-mediated dual targeting of stromal and immune components to overcome fibrotic and immunosuppressive barriers in hepatocellular carcinoma. J Control Release 2025; 383:113783. [PMID: 40306574 DOI: 10.1016/j.jconrel.2025.113783] [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: 03/03/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are key drivers of hepatocellular carcinoma (HCC) through their promotion of fibrosis and immune suppression activity. To overcome this stroma-immune barrier, we developed D/F@MRL, a stroma-immune co-targeting nanoplatform that enables the spatiotemporal coordination of CAF reprogramming and immune activation. In D/F@MRL, MMP-2-responsive hybrid liposomes (MRL) was employed to co-load digoxin (Dig) and PD-L1-degrading nanofibers (NFs). Upon encountering the MMP-2-enriched HCC stroma, D/F@MRL undergoes enzymatic cleavage, thereby enabling the targeted release of Dig and NFs within the HCC microenvironment. Mechanistically, Dig inhibits the phosphorylation of SMAD3 in CAFs, while PD-L1 degradation destabilizes the TGFβ receptor, synergistically silencing TGF-β/Smad signaling to reprogram CAFs. This combination not only disrupts the fibrotic barrier but also creates a feed-forward loop that further enhances drug penetration, while reinforcing the immune activation driven by Dig-induced immunogenic cell death (ICD) and PD-L1 degradation. In the humanized immune PDX model, D/F@MRL successfully reprogrammed CAFs and robustly remodeled the stromal and immune microenvironments without causing systemic toxicity, highlighting its promising potential for clinical translation. By integrating CAF reprogramming with ICD and immune checkpoint inhibition, this strategy overcame the limitations of single-target therapies, induced robust immune activation, further provided a clinic-transformative approach for fibrotic malignancies.
Collapse
Affiliation(s)
- Xiangyi Yin
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xingyang Zhao
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yiming Shen
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Weizhong Xie
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Cheng He
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jianan Guo
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zirong Li
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Feichao Xuan
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Silue Zeng
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiaojun Zeng
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chihua Fang
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Institute of Digital Intelligent Minimally Invasive Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou 510280, China; South China Institute of National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Guangzhou 510280, China.
| |
Collapse
|
23
|
Paál Á, Dora D, Takács Á, Rivard C, Pickard SL, Hirsch FR, Roskó B, Kiraly P, Ferdinandy P, Varga ZV, Lohinai Z, Görbe A. Roles of Annexin A1 Expression in Small Cell Lung Cancer. Cancers (Basel) 2025; 17:1407. [PMID: 40361334 PMCID: PMC12070913 DOI: 10.3390/cancers17091407] [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: 01/30/2025] [Revised: 03/21/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Small cell lung cancer (SCLC) is one of the malignancies with the worst prognosis, and there have been no major breakthroughs in its treatment for a long time. The majority of patients are diagnosed at the extensive stage, where the only option is chemotherapy, and even the addition of immune checkpoint inhibitors results in only modest benefits. The characterization of the molecular mechanisms behind therapy resistance has relevance in finding novel therapeutic approaches. Previous studies showed the possibility of annexin A1's (ANXA1) involvement in the immunosuppressive tumor microenvironment in SCLC, and there are studies showing the direct effects of ANXA1 modulation on cancer cell aggressiveness. METHODS We aimed to characterize the roles of ANXA1 expression using publicly available transcriptomic data, the RNA-seq-based predictive algorithms EPIC and ESTIMATE, and immunohistochemistry on patient samples. For the in vitro studies, we silenced ANXA1 expression with short hairpin RNA in three SCLC cell lines, measured the growth rate with the trypan blue exclusion assay, assessed the chemosensitivity to cisplatin and etoposide with the Presto BlueTM viability assay, and performed Western blots to assess changes in the levels of metabolic and mesenchymal markers and transcriptional drivers. RESULTS ANXA1-high tumors are associated with significantly increased immune infiltrates, stromality, and tumor-associated macrophages (TAMs). The ANXA1 protein is expressed on tumor cells and TAMs at the tissue level. ANXA1 silencing in H841 cells did not affect the growth rate; in SW1271 cells, shANXA1 cells grew significantly slower than shCTRL cells. Meanwhile, in H1048 cells, proliferation was significantly faster. Despite the different growth rates of the tested cell lines, ANXA1 silencing decreased the chemosensitivity to both cisplatin and etoposide in all three cell lines. Gene expression changes in mesenchymal markers, metabolic markers, dominant transcriptional drivers, and immune-relevant molecules were also characterized. CONCLUSIONS This is the first comprehensive characterization of ANXA1 in SCLC to reveal its role in the tumor's cell biology and the TME, aiming to boost further research in the field.
Collapse
Affiliation(s)
- Ágnes Paál
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and Pharmacotherapy, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, 1089 Budapest, Hungary
| | - David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, 1085 Budapest, Hungary; (D.D.); (B.R.)
| | - Ákos Takács
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
| | - Christopher Rivard
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.R.); (S.L.P.); (F.R.H.)
| | - Shivaun Lueke Pickard
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.R.); (S.L.P.); (F.R.H.)
| | - Fred R. Hirsch
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.R.); (S.L.P.); (F.R.H.)
- Tisch Cancer Institute, Center for Thoracic Oncology, Mount Sinai Health System, New York, NY 10029, USA
| | - Brigitta Roskó
- Department of Anatomy, Histology and Embryology, Semmelweis University, 1085 Budapest, Hungary; (D.D.); (B.R.)
| | - Peter Kiraly
- Translational Medicine Institute, Semmelweis University, 1085 Budapest, Hungary;
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Zoltán V. Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and Pharmacotherapy, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, 1089 Budapest, Hungary
| | - Zoltan Lohinai
- Translational Medicine Institute, Semmelweis University, 1085 Budapest, Hungary;
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
| |
Collapse
|
24
|
Laga T, Van Rompuy AS, Busschaert P, Marquina G, Loverix L, Olbrecht S, Ottenbourgs T, Baert T, Van Gorp T, Vergote I, Lambrechts D, Van Nieuwenhuysen E. Single-cell profiling in ovarian germ cell and sex cord-stromal tumours. Br J Cancer 2025:10.1038/s41416-025-03012-6. [PMID: 40269311 DOI: 10.1038/s41416-025-03012-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND The tumour microenvironment of rare ovarian germ cell tumours (OGCT) and sex-cord stromal tumours (SCST) remains unexplored. To better understand their immune and stromal landscape, we constructed a blueprint using single-cell RNA sequencing (scRNA-seq). METHODS We performed scRNA-seq of 66, 919 cells from twelve fresh tumour samples: seven adult granulosa cell tumour (aGSCT), one juvenile GSCT (jGSCT), one Sertoli-Leydig (SL) tumour, two immature teratoma (IT) and one dysgerminoma (DG). We characterised immune cell subtypes and fibroblasts based on their specific marker genes. Validation included combined positive score (CPS) of 46 OGCTs and 66 SCSTs, and bulk RNA sequencing (n = 32). RESULTS Cell clustering and annotation revealed a immune-activated microenvironment in DG, driven by PD-1- exhausted T cells, reflected in high CPS (≥10) and upregulated immune pathways. IT samples displayed no immunoreactive profile, consistent with a negative CPS. aGSCTs exhibited a fibroblast-enriched, immune-desert phenotype, with low T cell infiltration and increased immunosuppressive LYVE1 and CX3CR1+ macrophages, corresponding to negative CPS. CONCLUSION We constructed a detailed blueprint of the OGCT and SCSTs microenvironment of, elucidating potential modulators that shape their immune landscape. The immune-suppressive environment in aGSCTs likely limits immunotherapy response, as immunosuppressive macrophages inhibit T cell expansion along with EMT activation and fibroblast predominance.
Collapse
Affiliation(s)
- T Laga
- Department of Gynaecological Oncology, University Hospital Leuven, Leuven, Belgium.
- Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium.
- VIB Centre for Cancer Biology, Lab of Translational Genetics, Leuven, Belgium.
| | - A S Van Rompuy
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
- Laboratory of Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven-University of Leuven, Leuven, Belgium
| | - P Busschaert
- Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
- VIB Centre for Cancer Biology, Lab of Translational Genetics, Leuven, Belgium
| | - G Marquina
- Department of Medical Oncology, San Carlos Hospital, Madrid, Spain
| | - L Loverix
- Department of Gynaecological Oncology, University Hospital Leuven, Leuven, Belgium
| | - S Olbrecht
- Department of Gynaecological Oncology, University Hospital Leuven, Leuven, Belgium
| | - T Ottenbourgs
- Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
| | - T Baert
- Department of Gynaecological Oncology, University Hospital Leuven, Leuven, Belgium
| | - T Van Gorp
- Department of Gynaecological Oncology, University Hospital Leuven, Leuven, Belgium
- Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
| | - I Vergote
- Department of Gynaecological Oncology, University Hospital Leuven, Leuven, Belgium
| | - D Lambrechts
- VIB Centre for Cancer Biology, Lab of Translational Genetics, Leuven, Belgium
| | - E Van Nieuwenhuysen
- Department of Gynaecological Oncology, University Hospital Leuven, Leuven, Belgium
- Laboratory of Gynaecological Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
25
|
Weber F, Reese KL, Pantel K, Smit DJ. Cancer-associated fibroblasts as a potential novel liquid biopsy marker in cancer patients. J Exp Clin Cancer Res 2025; 44:127. [PMID: 40259388 PMCID: PMC12010557 DOI: 10.1186/s13046-025-03387-7] [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: 01/14/2025] [Accepted: 04/07/2025] [Indexed: 04/23/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are tissue residing cells within the tumor microenvironment (TME). Stromal CAFs have been shown to be associated with poor prognosis and tumor progression in several solid tumor entities. Although the molecular mechanisms are not fully understood yet, a critical role within the TME through direct interaction with the tumor cells as well as other cells has been proposed. While most studies on CAFs focus on stromal CAFs, recent reports highlight the possibility of detecting circulating CAFs (cCAFs) in the blood. In contrast to invasive tissue biopsies for stromal CAF characterization, liquid biopsy allows a minimally invasive isolation of cCAFs. Furthermore, liquid biopsy methods could enable continuous monitoring of cCAFs in cancer patients and therefore may present a novel biomarker for solid tumors. In this work, we present an overview of cCAF studies currently available and summarize the liquid biopsy techniques for cCAF isolation and detection. Moreover, the future research directions in the emerging field are highlighted and the potential applications of cCAFs as novel biomarkers for solid tumor patients discussed.
Collapse
Affiliation(s)
- Franziska Weber
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Kim-Lea Reese
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- European Liquid Biopsy Society (ELBS), Martinistraße 52, 20246, Hamburg, Germany
| | - Daniel J Smit
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
26
|
Kalfon J, Samaran J, Peyré G, Cantini L. scPRINT: pre-training on 50 million cells allows robust gene network predictions. Nat Commun 2025; 16:3607. [PMID: 40240364 PMCID: PMC12003772 DOI: 10.1038/s41467-025-58699-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
A cell is governed by the interaction of myriads of macromolecules. Inferring such a network of interactions has remained an elusive milestone in cellular biology. Building on recent advances in large foundation models and their ability to learn without supervision, we present scPRINT, a large cell model for the inference of gene networks pre-trained on more than 50 million cells from the cellxgene database. Using innovative pretraining tasks and model architecture, scPRINT pushes large transformer models towards more interpretability and usability when uncovering the complex biology of the cell. Based on our atlas-level benchmarks, scPRINT demonstrates superior performance in gene network inference to the state of the art, as well as competitive zero-shot abilities in denoising, batch effect correction, and cell label prediction. On an atlas of benign prostatic hyperplasia, scPRINT highlights the profound connections between ion exchange, senescence, and chronic inflammation.
Collapse
Affiliation(s)
- Jérémie Kalfon
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Machine Learning for Integrative Genomics group, F-75015, Paris, France
| | - Jules Samaran
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Machine Learning for Integrative Genomics group, F-75015, Paris, France
| | - Gabriel Peyré
- CNRS and DMA de l'Ecole Normale Supérieure, CNRS, Ecole Normale Supérieure, Université PSL, 75005, Paris, France
| | - Laura Cantini
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Machine Learning for Integrative Genomics group, F-75015, Paris, France.
| |
Collapse
|
27
|
Kim EY, Abides J, Keller CR, Martinez SR, Li W. Tumor Microenvironment Lactate: Is It a Cancer Progression Marker, Immunosuppressant, and Therapeutic Target? Molecules 2025; 30:1763. [PMID: 40333742 PMCID: PMC12029365 DOI: 10.3390/molecules30081763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/12/2025] [Accepted: 04/12/2025] [Indexed: 05/09/2025] Open
Abstract
The "Warburg effect" is a term coined a century ago for the preferential use of glycolysis over aerobic respiration in tumor cells for energy production, even under aerobic conditions. Although this is a less efficient mechanism of generating energy from glucose, aerobic glycolysis, in addition to the canonical anaerobic glycolysis, is an effective means of lactate production. The abundant waste product, lactate, yielded by the dual glycolysis in a tumor, has been discovered to be a major biomolecule that drives cancer progression. Lactate is a metabolic energy source that, via cell membrane lactate transporters, shuttles in and out of cancer cells as well as cancer cell-associated stromal cells and immune cells within the tumor microenvironment (TME). Additionally, lactate serves as a pH tuner, signaling ligand and transducer, epigenetic and gene transcription regulator, TME modifier, immune suppressor, chemoresistance modulator, and prognostic marker. With such broad functionalities, the production-consumption-reproduction of TME lactate fuels tumor growth and dissemination. Here, we elaborate on the lactate sources that contribute to the pool of lactate in the TME, the functions of TME lactate, the influence of the TME lactate on immune cell function and local tissue immunity, and anticancer therapeutic approaches adopting lactate manipulations and their efficacies. By scrutinizing these properties of the TME lactate and others that have been well addressed in the field, it is expected that a better weighing of the influence of the TME lactate on cancer development, progression, prognosis, and therapeutic efficacy can be achieved.
Collapse
Affiliation(s)
- Eugene Y. Kim
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (E.Y.K.); (J.A.); (C.R.K.)
| | - Joyce Abides
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (E.Y.K.); (J.A.); (C.R.K.)
- Doctor of Medicine Program, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| | - Chandler R. Keller
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (E.Y.K.); (J.A.); (C.R.K.)
| | - Steve R. Martinez
- Department of Medical Education and Clinical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
- Providence Regional Cancer Partnership, Providence Regional Medical Center, Everett, WA 98201, USA
| | - Weimin Li
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (E.Y.K.); (J.A.); (C.R.K.)
| |
Collapse
|
28
|
Ramos C, Walterskirchen N, Knöbl V, Zotter C, Müller C, Gerakopoulos V, Rauch A, Falk L, Sachet M, D'Angelo E, Agostini M, Pils D, Aust S, Grusch M, Herzog R, Kratochwill K, Le Blanc S, Lenos KJ, Vermeulen L, Riss S, Bachleitner-Hofmann T, Strobel O, Dolznig H, Bergmann M, Brostjan C, Unger LW, Oehler R. Colorectal cancer peritoneal metastasis is promoted by tissue-specific fibroblasts that can arise in response to various local disorders. Cancer Lett 2025:217686. [PMID: 40228602 DOI: 10.1016/j.canlet.2025.217686] [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: 12/02/2024] [Revised: 03/05/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025]
Abstract
Peritoneal membrane injury induces the activation of local fibroblasts and tissue remodelling, which ultimately can progress to fibrosis. Metastasis of colorectal cancer (CRC) to the abdominal cavity results in such peritoneal damage. Patients with colorectal cancer peritoneal metastasis (CPM) have a particularly poor prognosis, and CPM tumours are characterised by a high infiltration of fibroblasts. Here, we characterised the molecular and functional features of these fibroblasts, and investigated their interaction with other cells in the peritoneal microenvironment. Primary fibroblasts were isolated from 89 patients with different malignant and benign disorders of the peritoneum. We performed comprehensive analyses of single-cell and transcriptome profiling, secretome characterization, and functional enzymatic activity. We were able to identify a peritoneum-specific fibroblast population that increases in response to different types of damage-inducing peritoneal pathologies, including metastasis. These fibroblasts are characterised by the IGFBP2-dependent expression of CD38, which mediates extracellular non-canonical adenosinergic activity and contributes to the suppression of macrophages and T cells. Importantly, peritoneal fibroblasts promoted the growth and invasiveness of tumour cells in a xenograft mouse model of peritoneal metastasis, highlighting their pro-tumorigenic role. Their specific gene signature was associated with poor prognosis in a dataset of 51 patients suffering from colorectal peritoneal metastasis. This study revealed that the CPM is infiltrated by a peritoneal fibroblast subtype, which is absent in healthy tissue, but also observed in benign peritoneal diseases. Given the limited therapeutic options for these patients, these pro-tumorigenic peritoneal fibroblasts could represent an attractive target for inhibiting the peritoneal spread of tumour cells.
Collapse
Affiliation(s)
- Cristiano Ramos
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Natalie Walterskirchen
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Viktoria Knöbl
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Chiara Zotter
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Catharina Müller
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Vasileios Gerakopoulos
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Anna Rauch
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Lena Falk
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Monika Sachet
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Edoardo D'Angelo
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128, Padua, Italy
| | - Marco Agostini
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128, Padua, Italy
| | - Dietmar Pils
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Stefanie Aust
- Department of Obstetrics and Gynaecology, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael Grusch
- Center for Cancer Research, Medical University of Vienna, 1090, Vienna, Austria
| | - Rebecca Herzog
- Division of Paediatric Nephrology and Gastroenterology, Department of Paediatrics and Adolescent Medicine, Comprehensive Center for Paediatrics, Medical University of Vienna, 1090, Vienna, Austria
| | - Klaus Kratochwill
- Division of Paediatric Nephrology and Gastroenterology, Department of Paediatrics and Adolescent Medicine, Comprehensive Center for Paediatrics, Medical University of Vienna, 1090, Vienna, Austria
| | - Solange Le Blanc
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Kristiaan J Lenos
- Laboratory of Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC and University of Amsterdam, 1081 BT, Amsterdam, the Netherlands
| | - Louis Vermeulen
- Laboratory of Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC and University of Amsterdam, 1081 BT, Amsterdam, the Netherlands
| | - Stefan Riss
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Thomas Bachleitner-Hofmann
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Oliver Strobel
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Helmut Dolznig
- Institute of Medical Genetics, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael Bergmann
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Christine Brostjan
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Lukas W Unger
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria; Department of Colorectal Surgery, Oxford University Hospitals, Old Rd, Headington, Oxford, OX3 7LE, United Kingdom
| | - Rudolf Oehler
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
29
|
Ghazimoradi MH, Babashah S. The transcriptional regulators GATA6 and TET1 regulate the TGF-β pathway in cancer-associated fibroblasts to promote breast cancer progression. Cell Death Discov 2025; 11:164. [PMID: 40216762 PMCID: PMC11992015 DOI: 10.1038/s41420-025-02438-4] [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: 12/09/2024] [Revised: 03/13/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are pivotal drivers of tumor progression, yet the molecular mechanisms underlying their activation remain incompletely understood. Here, we identified the TET1/SMAD4/GATA6 regulatory axis as a central mechanism governing CAF transformation and function in breast cancer. Through integrative in vitro and in vivo models, we demonstrated that TET1, an epigenetic modulator, demethylates the SMAD4 promoter, enhancing SMAD4 expression. SMAD4 transcriptionally upregulates GATA6, which amplifies TGF-β signaling by directly activating the TGF-β promoter, establishing a self-reinforcing feedforward loop critical for CAF identity and stromal-tumor crosstalk. GATA6 and TET1 were significantly upregulated in breast CAFs compared to normal fibroblasts (NFs) and TGF-β-induced CAFs. Loss- or gain-of-function experiments revealed that these regulators control CAF survival, marker expression, and secretion of pro-tumorigenic factors. Knockdown of GATA6 or TET1 reduced CAF-mediated migration and invasion of breast cancer cells in vitro, while their overexpression enhanced cancer cell aggressiveness. Mechanistically, TET1-mediated epigenetic remodeling and GATA6-driven transcriptional activation converge on the TGF-β/SMAD pathway, sustaining CAF activation. In vivo, tumors derived from GATA6- or TET1-depleted CAFs exhibited reduced growth, proliferation, and CAF engraftment, underscoring their role in tumor progression. These findings position GATA6 and TET1 as promising targets to disrupt CAF-driven tumorigenesis, offering novel strategies for breast cancer treatment. By unraveling the epigenetic-transcriptional interplay within the tumor microenvironment, this study advances our understanding of stromal reprogramming and its implications for precision oncology.
Collapse
Affiliation(s)
- Mohammad H Ghazimoradi
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
30
|
Katsumata H, Koguchi D, Hirano S, Suzuki A, Yanagita K, Shimizu Y, Hirono W, Shimura S, Ikeda M, Tsumura H, Ishii D, Sato Y, Matsumoto K. Association Between CKAP4 Expression and Poor Prognosis in Patients with Bladder Cancer Treated with Radical Cystectomy. Cancers (Basel) 2025; 17:1278. [PMID: 40282454 PMCID: PMC12025835 DOI: 10.3390/cancers17081278] [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: 03/11/2025] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: While cytoskeleton-associated protein 4 (CKAP4) has been associated with prognosis in various malignancies, its prognostic value for bladder cancer (BCa) remains unclear. The aim of this study was to evaluate CKAP4 expression in tumor cells and cancer-associated fibroblasts (CAFs) following radical cystectomy (RC) in patients with BCa. Methods: In this study, CKAP4 in tumor cells was defined as CKAP4-1, while CKAP4 expressed in CAFs was defined as CKAP4-2. CKAP4-2 expression was evaluated to explore its potential association with tumor aggressiveness and patient outcomes. CKAP4 expression in 86 RC specimens was assessed using immunohistochemistry. CKAP4-1 positivity was considered when ≥5% cytoplasmic staining of cancer cells, with at least moderate staining intensity, was observed. CKAP4-2 positivity was evaluated using a point scale (0-3), with scores based on the number of CKAP4 positive CAFs in the tumor stroma. Scores of 2 (moderate number of CAFs) and 3 (significant number of CAFs) were considered to indicate positivity. Results: CKAP4-1 and CKAP4-2 were expressed in 53 (61.6%) and 34 (39.5%) patients, respectively. Kaplan-Meier analysis showed that patients with CKAP4-1 had significantly shorter cancer-specific survival and recurrence-free survival (RFS; p = 0.046 and p = 0.0173, respectively). Multivariate analysis showed that CKAP4-1 positivity was an independent predictor of RFS (p = 0.041, hazard ratio: 2.09, 95% confidence interval: 1.03-4.25). Conclusions: This study showed that CKAP4 expression in tumor cells may serve as a useful prognostic biomarker for patients with BCa who undergo RC.
Collapse
Affiliation(s)
- Hiroki Katsumata
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara 252-0374, Japan; (H.K.)
| | - Dai Koguchi
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara 252-0374, Japan; (H.K.)
| | - Shuhei Hirano
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara 252-0374, Japan; (H.K.)
| | - Anna Suzuki
- Department of Pathology, Nagaoka Chuo General Hospital, 2041 Kawasaki, Nagaoka 940-0861, Japan
| | - Kengo Yanagita
- Biofluid Biomarker Center, Niigata University, 8050 ikarashi 2-no-cho Nishi-ku, Niigata 950-2181, Japan
| | - Yuriko Shimizu
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara 252-0374, Japan; (H.K.)
| | - Wakana Hirono
- Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara 252-0374, Japan
| | - Soichiro Shimura
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara 252-0374, Japan; (H.K.)
| | - Masaomi Ikeda
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara 252-0374, Japan; (H.K.)
| | - Hideyasu Tsumura
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara 252-0374, Japan; (H.K.)
| | - Daisuke Ishii
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara 252-0374, Japan; (H.K.)
| | - Yuichi Sato
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara 252-0374, Japan; (H.K.)
- KITASATO-OTSUKA Biomedical Assay Laboratories Co., Ltd., 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0329, Japan
| | - Kazumasa Matsumoto
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara 252-0374, Japan; (H.K.)
| |
Collapse
|
31
|
Zhu X, Li B, Qin L, Liang T, Hu W, Li J, Wang J. Development and Validation of a Prognostic Model for Lung Adenocarcinoma Based on CAF-Related Genes: Unveiling the Role of COX6A1 in Cancer Progression and CAF Infiltration. Int J Mol Sci 2025; 26:3478. [PMID: 40331946 PMCID: PMC12026577 DOI: 10.3390/ijms26083478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Lung adenocarcinoma (LUAD), the predominant subtype of non-small cell lung cancer (NSCLC), presents significant challenges in early diagnosis and personalized treatment. Recent research has focused on the role of the tumor microenvironment, particularly tumor-associated fibroblasts (CAFs), in tumor progression. This study systematically analyzed CAF immune infiltration-related genes to construct a prognostic model for LUAD, confirming its predictive value for patient outcomes. The risk score derived from CAF-related genes (CAFRGs) was negatively correlated with immune microenvironment scores and linked to the expression of immune checkpoint genes, indicating that high-risk patients may exhibit immune escape characteristics. Analysis via the TIDE tool revealed that low-risk patients had more active T-cell immune responses. The risk score also correlated with anti-tumor drug sensitivity, particularly to doramapimod. Notably, COX6A1 emerged as a key gene in the model, with its upregulation associated with immune cell infiltration and immune escape. Further in vitro experiments demonstrated that COX6A1 regulates LUAD cell migration, proliferation, and senescence, suggesting its role in tumor immune evasion. Additionally, further co-culture studies of lung cancer cells and fibroblasts revealed that COX6A1 knockdown promotes the expression of CAF-related cytokines, enhancing CAF infiltration. Overall, this study provides a foundation for personalized treatment of LUAD and highlights COX6A1 as a promising therapeutic target within the tumor immune microenvironment, guiding future clinical research.
Collapse
Affiliation(s)
| | | | | | | | | | - Jianxiang Li
- School of Public Health, Suzhou Medicine College of Soochow University, Suzhou 215123, China; (X.Z.); (B.L.); (L.Q.); (T.L.); (W.H.)
| | - Jin Wang
- School of Public Health, Suzhou Medicine College of Soochow University, Suzhou 215123, China; (X.Z.); (B.L.); (L.Q.); (T.L.); (W.H.)
| |
Collapse
|
32
|
Liu K, Cui Y, Han H, Guo E, Shi X, Xiong K, Zhang N, Zhai S, Sang S, Liu M, Chen B, Gu Y. Fibroblast atlas: Shared and specific cell types across tissues. SCIENCE ADVANCES 2025; 11:eado0173. [PMID: 40173240 PMCID: PMC11963979 DOI: 10.1126/sciadv.ado0173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/26/2025] [Indexed: 04/04/2025]
Abstract
Understanding the heterogeneity of fibroblasts depends on decoding the complexity of cell subtypes, their origin, distribution, and interactions with other cells. Here, we integrated 249,156 fibroblasts from 73 studies across 10 tissues to present a single-cell atlas of fibroblasts. We provided a high-resolution classification of 18 fibroblast subtypes. In particular, we revealed a previously undescribed cell population, TSPAN8+ chromatin remodeling fibroblasts, characterized by high expression of genes with functions related to histone modification and chromatin remodeling. Moreover, TSPAN8+ chromatin remodeling fibroblasts were detectable in spatial transcriptome data and multiplexed immunofluorescence assays. Compared with other fibroblast subtypes, TSPAN8+ chromatin remodeling fibroblasts exhibited higher scores in cell differentiation and resident fibroblast, mainly interacting with endothelial cells and T cells through ligand VEGFA and receptor F2R, and their presence was associated with poor prognosis. Our analyses comprehensively defined the shared and specific characteristics of fibroblast subtypes across tissues and provided a user-friendly data portal, Fibroblast Atlas.
Collapse
Affiliation(s)
- Kaidong Liu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yanrui Cui
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Huiming Han
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Erliang Guo
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Xingyang Shi
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Kai Xiong
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Nan Zhang
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Songmei Zhai
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shaocong Sang
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Mingyue Liu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Bo Chen
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yunyan Gu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
33
|
Ma S, Lu Y, Sui S, Yang JS, Fu BB, Tan PX, Chai Y, Lv J, Kong L, Wu X, Gao YB, Yan T. Unraveling the triad of immunotherapy, tumor microenvironment, and skeletal muscle biomechanics in oncology. Front Immunol 2025; 16:1572821. [PMID: 40242775 PMCID: PMC12000078 DOI: 10.3389/fimmu.2025.1572821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/03/2025] [Indexed: 04/18/2025] Open
Abstract
The intricate interaction between skeletal muscle biomechanics, the tumor microenvironment, and immunotherapy constitutes a pivotal research focus oncology. This work provides a comprehensive review of methodologies for evaluating skeletal muscle biomechanics, including handheld dynamometry, advanced imaging techniques, electrical impedance myography, elastography, and single-fiber experiments to assess muscle quality and performance. Furthermore, it elucidates the mechanisms, applications, and limitations of various immunotherapy modalities, including immune checkpoint inhibitors, adoptive cell therapy, cancer vaccines, and combined chemoimmunotherapy, while examining their effects on skeletal muscle function and systemic immune responses. Key findings indicate that although immunotherapy is effective in augmenting antitumor immunity, it frequently induces muscle-related adverse effects such as weakness, fatigue, or damage, primarily mediated by cytokine release and immune activation. This work underscores the significance of immune niches within the tumor microenvironment in influencing treatment outcomes and proposes strategies to optimize therapy through personalized regimens and combinatorial approaches. This review highlights the need for further research on the formation of immune niches and interactions muscle-tumor. Our work is crucial for advancing the efficacy of immunotherapy, reducing adverse effects, and ultimately improving survival rates and quality of life of patients with cancer.
Collapse
Affiliation(s)
- Shuang Ma
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Ying Lu
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Shang Sui
- St. John’s Kilmarnock School, Breslau, ON, Canada
| | - Jia-shuo Yang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bing-bing Fu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei-xin Tan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicheng Chai
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Jiaqi Lv
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Lingyu Kong
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Xiaolin Wu
- School of Mathematics and Statistics, Liaoning University, Shenyang, China
| | - Yi-bo Gao
- Department of Oral and Maxillofacial Surgery, Taikang Bybo Dental, Beijing, China
| | - Tao Yan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
34
|
Caloian AD, Cristian M, Calin E, Pricop AR, Mociu SI, Seicaru L, Deacu S, Ciufu N, Suceveanu AI, Suceveanu AP, Mazilu L. Epigenetic Symphony in Diffuse Large B-Cell Lymphoma: Orchestrating the Tumor Microenvironment. Biomedicines 2025; 13:853. [PMID: 40299416 PMCID: PMC12024808 DOI: 10.3390/biomedicines13040853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/30/2025] Open
Abstract
DLBCL is a testament to the complexity of nature. It is characterized by remarkable diversity in its molecular and pathological subtypes and clinical manifestations. Despite the strides made in DLBCL treatment and the introduction of innovative drugs, around one-third of patients face a relapse or develop refractory disease. Recent findings over the past ten years have highlighted the critical interplay between the evolution of DLBCL and various epigenetic mechanisms, including chromatin remodeling, DNA methylation, histone modifications, and the regulatory roles of non-coding RNAs. These epigenetic alterations are integral to the pathways of oncogenesis, tumor progression, and the development of therapeutic resistance. In the past decade, the identification of dysregulated epigenetic mechanisms in lymphomas has paved the way for an exciting field of epigenetic therapies. Crucially, these epigenetic transformations span beyond tumor cells to include the sophisticated network within the tumor microenvironment (TME). While the exploration of epigenetic dysregulation in lymphoma cells is thriving, the mechanisms affecting the functions of immune cells in the TME invite further investigation. This review is dedicated to weaving together the narrative of epigenetic alterations impacting both lymphoma cells with a focus on their infiltrating immune companions.
Collapse
Affiliation(s)
- Andreea-Daniela Caloian
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Department of Hemato-Oncology, “Ovidius” Clinical Hospital, 900470 Constanta, Romania;
| | - Miruna Cristian
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology-CEDMOG, “Ovidius” University of Constanta, 900470 Constanta, Romania
- Department of Forensic Medicine, “Sf. Apostol Andrei” Emergency County Hospital, 900439 Constanta, Romania
| | - Elena Calin
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Department of Hemato-Oncology, “Ovidius” Clinical Hospital, 900470 Constanta, Romania;
| | - Andreea-Raluca Pricop
- Department of Dermatology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania;
| | - Stelian-Ilie Mociu
- Department of Hemato-Oncology, “Ovidius” Clinical Hospital, 900470 Constanta, Romania;
| | - Liliana Seicaru
- Department of Clinical Patology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania;
| | - Sorin Deacu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Department of Clinical Patology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania;
| | - Nicolae Ciufu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Department of Hemato-Oncology, “Ovidius” Clinical Hospital, 900470 Constanta, Romania;
| | - Andra-Iulia Suceveanu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Department of Gastroenterology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
| | - Adrian-Paul Suceveanu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Department of Gastroenterology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
| | - Laura Mazilu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Department of Hemato-Oncology, “Ovidius” Clinical Hospital, 900470 Constanta, Romania;
| |
Collapse
|
35
|
Fatima S. Tumor Microenvironment: A Complex Landscape of Cancer Development and Drug Resistance. Cureus 2025; 17:e82090. [PMID: 40351953 PMCID: PMC12066109 DOI: 10.7759/cureus.82090] [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] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Cancer is responsible for nearly one in six global fatalities, making it a major health issue worldwide. Despite advancements in early detection, surgery, and targeted therapies, effective treatment remains challenging due to the complexity and heterogeneity of the disease. A key factor in cancer progression and resistance to treatment is the tumor microenvironment (TME). It is a complex ecosystem comprising cancer cells, stromal cells, immune cells, extracellular matrix (ECM), and soluble factors like cytokines and chemokines. These components interact dynamically to influence tumor growth, metastasis, immune evasion, and treatment resistance. Cancer cells drive the formation of the TME by releasing signaling molecules, while stromal cells, such as fibroblasts and endothelial cells, support tumor metabolism, angiogenesis, and invasion. Immune cells within the TME can either suppress or promote tumor progression, depending on their activation state. Additionally, the TME can promote the growth of immunosuppressive cells that aid cancer cells in evading immune surveillance, such as regulatory T-cells and myeloid-derived suppressor cells. The TME also impedes drug delivery by creating defective blood vessels, contributing to drug resistance. Recent technological advancements have deepened our understanding of the TME, revealing its role in immune modulation, metabolism, and extracellular matrix remodeling. As a result, targeting the TME has become a promising strategy to overcome treatment resistance and improve cancer therapy outcomes.
Collapse
Affiliation(s)
- Sohaila Fatima
- Pathology, College of Medicine, King Khalid University, Abha, SAU
| |
Collapse
|
36
|
Harada Y. Manipulating mannose metabolism as a potential anticancer strategy. FEBS J 2025; 292:1505-1519. [PMID: 39128015 DOI: 10.1111/febs.17230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/12/2024] [Accepted: 07/18/2024] [Indexed: 08/13/2024]
Abstract
Cancer cells acquire metabolic advantages over their normal counterparts regarding the use of nutrients for sustained cell proliferation and cell survival in the tumor microenvironment. Notable among the metabolic traits in cancer cells is the Warburg effect, which is a reprogrammed form of glycolysis that favors the rapid generation of ATP from glucose and the production of biological macromolecules by diverting glucose into various metabolic intermediates. Meanwhile, mannose, which is the C-2 epimer of glucose, has the ability to dampen the Warburg effect, resulting in slow-cycling cancer cells that are highly susceptible to chemotherapy. This anticancer effect of mannose appears when its catabolism is compromised in cancer cells. Moreover, de novo synthesis of mannose within cancer cells has also been identified as a potential target for enhancing chemosensitivity through targeting glycosylation pathways. The underlying mechanisms by which alterations in mannose metabolism induce cancer cell vulnerability are just beginning to emerge. This review summarizes the current state of our knowledge of mannose metabolism and provides insights into its manipulation as a potential anticancer strategy.
Collapse
Affiliation(s)
- Yoichiro Harada
- Department of Glyco-Oncology and Medical Biochemistry, Research Institute, Osaka International Cancer Institute, Japan
| |
Collapse
|
37
|
Liu H, Yong T, Zhang X, Wei Z, Bie N, Xu S, Zhang X, Li S, Zhang J, Zhou P, Yang X, Gan L. Spatial Regulation of Cancer-Associated Fibroblasts and Tumor Cells via pH-Responsive Bispecific Antibody Delivery for Enhanced Chemo-Immunotherapy Synergy. ACS NANO 2025; 19:11756-11773. [PMID: 40114589 DOI: 10.1021/acsnano.4c13277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
The effectiveness of chemotherapy is often compromised by physiological barriers and an immunosuppressive tumor microenvironment. Cancer-associated fibroblasts (CAFs) significantly contribute to the reconfiguration of the tumor extracellular matrix (ECM) and the suppression of immune responses, making them crucial targets for therapeutic intervention. Here, a tumor acidic microenvironment-responsive delivery system that utilizes tumor cell-derived microparticles (MPs) as carriers for the chemotherapeutic agent doxorubicin (DOX) and the bispecific antibody YM101 targeting both TGF-β and PD-L1 is developed (DOX@MPs-YM101) to spatially regulate both CAFs and tumor cells for enhanced chemotherapeutic efficacy. DOX@MPs-YM101 efficiently targets tumor tissues and releases DOX@MPs and YM101 in response to the acidic tumor microenvironment. YM101 reprograms CAFs and reduces the tumor ECM, facilitating tumor accumulation and deep penetration of DOX@MPs-YM101. DOX@MPs are highly internalized into tumor cells, triggering immunogenic cell death (ICD) and activating CD8+ T cell-mediated antitumor immunity. The reprogramming of CAFs by YM101 further promotes the accumulation of CD8+ T cells and reduces the number of immunosuppressive cells within the tumors. Additionally, YM101 effectively neutralizes PD-L1 on tumor cells induced by DOX@MPs, restoring CD8+ T cell activity and generating long-term antitumor immune memory to prevent tumor recurrence. Our findings highlight the potential of DOX@MPs-YM101 to improve chemotherapy in cancer treatment.
Collapse
Affiliation(s)
- Haojie Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaoqiong Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhaohan Wei
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Nana Bie
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shiyi Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaojuan Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shiyu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jing Zhang
- Wuhan YZY Biopharma Co., Ltd., Wuhan 430074, China
| | - Pengfei Zhou
- Wuhan YZY Biopharma Co., Ltd., Wuhan 430074, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
38
|
Ravi K, Zhang Y, Sakala L, Manoharan TJM, Pockaj B, LaBaer J, Park JG, Nikkhah M. Tumor Microenvironment On-A-Chip and Single-Cell Analysis Reveal Synergistic Stromal-Immune Crosstalk on Breast Cancer Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413457. [PMID: 40056038 PMCID: PMC12021108 DOI: 10.1002/advs.202413457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/11/2025] [Indexed: 04/26/2025]
Abstract
Solid tumors develop within a complex environment called the tumor microenvironment (TME), which is sculpted by the presence of other cells, such as cancer-associated fibroblasts (CAFs) and immune cells like macrophages (Mφs). Despite the presence of immune cells, tumor cells orchestrate a tumor-supportive environment through intricate interaction with the components of the TME. However, the specific mechanism by which this intercellular dialogue is regulated is not fully understood. To that end, the development of an organotypic 3D breast TME-on-a-chip (TMEC) model, integrated with single-cell RNA sequencing analysis, is reported to mechanistically evaluate the progression of triple-negative breast cancer (TNBC) cells in the presence of patient-derived CAFs and Mφs. Extensive functional assays, including invasion and morphometric characterization, reveal the synergistic influence of CAFs and Mφs on tumor cells. Furthermore, gene expression and pathway enrichment analyses identify the involvement of the KYNU gene, suggesting a potential immune evasion mechanism through the kynurenine pathway. Lastly, the pharmacological targeting of the identified pathway is investigated.
Collapse
Affiliation(s)
- Kalpana Ravi
- School of Biological and Health Systems Engineering (SBHSE)Arizona State UniversityTempeAZ85287USA
| | - Yining Zhang
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| | - Lydia Sakala
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| | | | | | - Joshua LaBaer
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| | - Jin G. Park
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE)Arizona State UniversityTempeAZ85287USA
- Biodesign Virginia G. Piper Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287USA
| |
Collapse
|
39
|
Ganbold M, Louphrasitthiphol P, Miyamoto T, Miyazaki Y, Oda T, Tominaga K, Isoda H. Isorhamnetin exerts anti-proliferative effect on cancer-associated fibroblasts by inducing cell cycle arrest. Biomed Pharmacother 2025; 185:117954. [PMID: 40031374 DOI: 10.1016/j.biopha.2025.117954] [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: 11/25/2024] [Revised: 02/16/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025] Open
Abstract
Isorhamnetin (ISO), a dietary flavonoid, has been shown to possess antioxidant, anti-cancer, and anti-inflammatory properties. Cancer-associated fibroblasts (CAFs), found in the tumor microenvironment of several types of cancer including pancreatic ductal adenocarcinoma (PDAC) impact the tumor growth and development of chemoresistance. Thus, modulating CAFs is an attractive mean to increase the efficacy of therapies targeting cancer cells. In this study, the anti-proliferative effect of ISO and the underlying transcriptomic profile of ISO-treated PDAC-derived CAFs were investigated. ISO treatment showed a time- and concentration-dependent decrease in cell viability with a slight increase in apoptotic cells. Microarray and cell cycle analyses revealed ISO induced downregulation of pathways in cell cycle and DNA replication; and G2/M checkpoint. Cell cycle analysis showed cells in the G2/M phase were increased. In response to the treatment, hallmark for p53 pathway genes, known to regulate cell cycle checkpoints, were highly upregulated. Moreover, ISO-treated cells had an increased area of the mitochondrial network, but lower mitochondrial membrane potential accompanied by a decrease of ATP production, measured by oxygen consumption rate. Inflammatory gene expression of IL1A1, IL6, CXCL1, and LIF were significantly inhibited in ISO-treated CAFs. Taken together, our results demonstrated that the cytostatic effect of ISO on human CAFs was mediated by inducing cell cycle arrest at G2/M phase associated with activation of p21, impaired mitochondrial homeostasis, and inhibition of inflammatory mediators gene expression, warranting further investigation for its use in combinatorial therapy that target both the cancer and the tumor microenvironment as a whole.
Collapse
Affiliation(s)
- Munkhzul Ganbold
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Science and Technology (AIST), Tsukuba, Japan
| | - Pakavarin Louphrasitthiphol
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan; Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford, UK
| | - Takafumi Miyamoto
- Department of Internal Medicine (Endocrinology and Metabolism), Institute of Medicine, University of Tsukuba, Tsukuba, Japan; Transborder Medical Research Center, University of Tsukuba, Tsukuba, Japan
| | - Yoshihiro Miyazaki
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tatsuya Oda
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kenichi Tominaga
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Science and Technology (AIST), Tsukuba, Japan
| | - Hiroko Isoda
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Science and Technology (AIST), Tsukuba, Japan; Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan; Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
40
|
Di Spirito A, Balkhi S, Vivona V, Mortara L. Key immune cells and their crosstalk in the tumor microenvironment of bladder cancer: insights for innovative therapies. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002304. [PMID: 40177538 PMCID: PMC11964778 DOI: 10.37349/etat.2025.1002304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Bladder cancer (BC) is a heterogeneous disease associated with high mortality if not diagnosed early. BC is classified into non-muscle-invasive BC (NMIBC) and muscle-invasive BC (MIBC), with MIBC linked to poor systemic therapy response and high recurrence rates. Current treatments include transurethral resection with Bacillus Calmette-Guérin (BCG) therapy for NMIBC and radical cystectomy with chemotherapy and/or immunotherapy for MIBC. The tumor microenvironment (TME) plays a critical role in cancer progression, metastasis, and therapeutic efficacy. A comprehensive understanding of the TME's complex interactions holds substantial translational significance for developing innovative treatments. The TME can contribute to therapeutic resistance, particularly in immune checkpoint inhibitor (ICI) therapies, where resistance arises from tumor-intrinsic changes or extrinsic TME factors. Recent advancements in immunotherapy highlight the importance of translational research to address these challenges. Strategies to overcome resistance focus on remodeling the TME to transform immunologically "cold" tumors, which lack immune cell infiltration, into "hot" tumors that respond better to immunotherapy. These strategies involve disrupting cancer-microenvironment interactions, inhibiting angiogenesis, and modulating immune components to enhance anti-tumor responses. Key mechanisms include cytokine involvement [e.g., interleukin-6 (IL-6)], phenotypic alterations in macrophages and natural killer (NK) cells, and the plasticity of cancer-associated fibroblasts (CAFs). Identifying potential therapeutic targets within the TME can improve outcomes for MIBC patients. This review emphasizes the TME's complexity and its impact on guiding novel therapeutic approaches, offering hope for better survival in MIBC.
Collapse
Affiliation(s)
- Anna Di Spirito
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Sahar Balkhi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Veronica Vivona
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
41
|
Brogna MR, Varone V, DelSesto M, Ferrara G. The role of CAFs in therapeutic resistance in triple negative breast cancer: an emerging challenge. Front Mol Biosci 2025; 12:1568865. [PMID: 40230452 PMCID: PMC11994926 DOI: 10.3389/fmolb.2025.1568865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/11/2025] [Indexed: 04/16/2025] Open
Abstract
The tumor microenvironment (TME) is a crucial element of cancerous tissue and has emerged as a promising target for therapeutic strategies. The complex variety of stromal cells within the TME plays a vital role in determining the tumor's aggressiveness and its resistance to treatment. Tumor progression is not solely driven by cancer cells harboring genetic mutations but is also significantly influenced by non-cancerous host cells within the TME, which strongly impact tumor growth, metastasis, and the response to therapies. Cancer-associated fibroblasts (CAFs) are a diverse group of stromal cells within the TME. They play dual roles, both promoting and inhibiting tumor growth, making them intriguing targets for enhancing cancer therapies. Their significant contribution to creating a tumor-supportive environment has diminished the effectiveness of various cancer treatments, including radiation, chemotherapy, immunotherapy, and hormone therapy. Research has increasingly focused on understanding how CAFs contribute to therapy resistance in triple-negative breast cancer (TNBC) to improve treatment outcomes. However, the ways in which CAF patterns affect the TME and the response to immunotherapy in TNBC are not yet well understood and the interactions between CAFs, tumor cells, and immune cells in TNBC remain largely unexplored. In this review, we thoroughly exam ine the relationship between TNBC progression and CAF patterns. We discuss the current understanding of CAF heterogeneity, their role in tumor progression, and their impact on the tumor's response to therapeutic agents in TNBC. Additionally, we explore the potential and possible strategies for therapies targeting CAFs.
Collapse
|
42
|
Luo Y, Zhang P, Wu Z, Zou L, Bai X, Li X, Gan W, Wang F, Han Z, Lin Q, Wang F, Gu Y. 99mTc-Labeled Quinolone-Based Novel Skeletal Tracers for Tumor Visualization through Fibroblast Activation Protein. J Med Chem 2025; 68:6735-6747. [PMID: 40085735 DOI: 10.1021/acs.jmedchem.5c00132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Fibroblast activation protein (FAP) has emerged as a prominent target for tumor diagnosis. Quinoline-based FAP PET tracers demonstrated clinical feasibility. However, there is a relative scarcity of clinical studies on 99mTc-labeled FAP SPECT tracers. The existing quinoline-derived 99mTc-FAPI tracer exhibits relatively low tumor uptake and suboptimal pharmacokinetic properties, which restrict its clinical application. Consequently, it is necessary to alter the pharmacophores to improve its druggability. In this study, a novel quinolone-based pharmacophore was developed by utilizing scaffold hopping and conformational constrained strategies. Serial screening and preclinical evaluations were conducted. The 99mTc-FAPI-YQ3 showed extremely high tumor uptake and excellent pharmacokinetic properties. Additionally, 99mTc-FAPI-YQ3 demonstrated reliable safety characteristics and clinical efficacy on four different oncology patients. In conclusion, 99mTc-FAPI-YQ3 was a promising radiotracer for FAP-targeted cancer diagnosis, shedding light on substantially advancing SPECT molecular imaging.
Collapse
Affiliation(s)
- Yang Luo
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Pengjun Zhang
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
- Department of Nuclear Medicine, Nanjing First Hospital, China Pharmaceutical University, Nanjing 210006, China
| | - Zihan Wu
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Lenan Zou
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Xuedong Bai
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Xue Li
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Wenhui Gan
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Faying Wang
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Zhihao Han
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Qiao Lin
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, China Pharmaceutical University, Nanjing 210006, China
| | - Yueqing Gu
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
43
|
Sun X, Cai W, Li H, Gao C, Ma X, Guo Y, Fu D, Xiao D, Zhang Z, Wang Y, Yang S, Feng Y, Zhao T, Hao J. Endothelial-like cancer-associated fibroblasts facilitate pancreatic cancer metastasis via vasculogenic mimicry and paracrine signalling. Gut 2025:gutjnl-2024-333638. [PMID: 40122596 DOI: 10.1136/gutjnl-2024-333638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 03/08/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are highly heterogeneous in the progression of pancreatic ductal adenocarcinoma (PDAC) and vasculogenic mimicry (VM) refers to a phenomenon in which cancer cells adopt endothelial-like characteristics. OBJECTIVE To identify a novel protumoural CAF subtype undertaking VM. DESIGN We used single-cell RNA sequencing and mIHC to identify FAPα+CD144+ endothelial-like CAFs (endoCAFs) and combined prospective and retrospective analyses to assess its clinical outcomes. Tube formation, proliferation and invasion assay were conducted on cell lines, organoids, the orthotopic tumour model and LSL-KrasG12D/+, LSL-Trp53R172H/+ and Pdx1-Cre (KPC) mouse model. Mechanically, we performed cytokine array assays, RNA-sequencing, IP-mass spectrometry, ChIP and luciferase analyses. Importantly, an siRNA delivery nanosystem was developed to precisely target FAPα+CD144+endoCAFs in vivo. RESULTS FAPα+CD144+endoCAFs were present in the tumour microenvironment of PDAC, and patients with a higher CD144+CAFs proportion displayed poor prognosis of PDAC. FAPα+CD144+endoCAFs not only acquired a VM phenotype to provide metastatic conduits but also promoted the proliferation and invasion of tumour cells in situ through paracrine signalling, thereby actively facilitating the metastasis of tumour cells. The CD144-β-catenin-STAT3 signalling axis was activated, and CD144 and downstream secreted cytokines were transcriptionally upregulated to maintain the dual roles of endoCAFs. A CAF-targeting siRNA delivery nanosystem, via loading FAPα and siCD144, was administered to precisely target FAPα+CD144+ endoCAFs, which substantially inhibited their protumoural roles in vivo. CONCLUSION FAPα+CD144+endoCAFs can promote metastasis of PDAC via undertaking VM and paracrine through activation of the CD144-β-catenin-STAT3 signalling axis. CAF-targeting siRNA delivery nanosystem can inhibit tumour progression by precisely targeting FAPα+CD144+endoCAFs.
Collapse
Affiliation(s)
- Xugang Sun
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Wenrun Cai
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Haorui Li
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Chuntao Gao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Xi Ma
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Yu Guo
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Danqi Fu
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Di Xiao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Zhaoyu Zhang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Yifei Wang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Shengyu Yang
- Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Yukuan Feng
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Tiansuo Zhao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Jihui Hao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| |
Collapse
|
44
|
Ma J, Tang L, Xiao J, Tang K, Zhang H, Huang B. Burning lactic acid: a road to revitalizing antitumor immunity. Front Med 2025:10.1007/s11684-025-1126-6. [PMID: 40119026 DOI: 10.1007/s11684-025-1126-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/16/2024] [Indexed: 03/24/2025]
Abstract
Lactic acid (LA) accumulation in tumor microenvironments (TME) has been implicated in immune suppression and tumor progress. Diverse roles of LA have been elucidated, including microenvironmental pH regulation, signal transduction, post-translational modification, and metabolic remodeling. This review summarizes LA functions within TME, focusing on the effects on tumor cells, immune cells, and stromal cells. Reducing LA levels is a potential strategy to attack cancer, which inevitably affects the physiological functions of normal tissues. Alternatively, transporting LA into the mitochondria as an energy source for immune cells is intriguing. We underscore the significance of LA in both tumor biology and immunology, proposing the burning of LA as a potential therapeutic approach to enhance antitumor immune responses.
Collapse
Affiliation(s)
- Jingwei Ma
- Department of Immunology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China.
| | - Liang Tang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Jingxuan Xiao
- Department of Immunology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China
| | - Ke Tang
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China
| | - Huafeng Zhang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bo Huang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China.
| |
Collapse
|
45
|
Zhang B, Zhang Z, Gao J, Lu S, Pang R, Li D, Huang X, Qin N, Liu L, Wang Z. Targeting FAK improves the tumor uptake of antibody-drug conjugates to strengthen the anti-cancer responses. iScience 2025; 28:111536. [PMID: 40040813 PMCID: PMC11879607 DOI: 10.1016/j.isci.2024.111536] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/04/2024] [Accepted: 12/03/2024] [Indexed: 03/06/2025] Open
Abstract
Antibody-drug conjugates (ADCs), exemplified by HER2-targeted Enhertu and TROP2-targeted Trodelvy, have demonstrated significant therapeutic potential in cancers. However, a subset of patients remains refractory to ADC treatment, suggesting that the efficacy requires further optimization. Here, we demonstrate that excessive cancer-associated fibroblasts (CAFs) can form a fibrotic barrier, impeding the tissue uptake of ADCs to dampen the anti-tumor efficacy. Mechanistically, cancer cells transform normal fibroblasts into FAK-activated CAFs. The proliferation of these CAFs reduces the tumor uptake of macromolecular drugs, conferring resistance to ADCs. Targeting FAK with a small molecule inhibitor IN10018 effectively diminishes the CAF-associated tumor barrier, enhancing the tumor uptake of various ADCs irrespective of their specific targets. Combination therapy with IN10018 and ADCs targeting either HER2 or TROP2 consistently yielded superior antitumor outcomes compared to monotherapies in animal models. These findings provide compelling preclinical evidence supporting the clinical evaluation of IN10018 in combination with ADCs.
Collapse
Affiliation(s)
| | | | - Jiaming Gao
- InxMed (Shanghai) Co., Ltd., Shanghai, China
| | - Shiqiang Lu
- InxMed (Shanghai) Co., Ltd., Shanghai, China
| | - Ran Pang
- InxMed (Shanghai) Co., Ltd., Shanghai, China
| | | | | | - Natasha Qin
- InxMed (Shanghai) Co., Ltd., Shanghai, China
| | - Leo Liu
- InxMed (Shanghai) Co., Ltd., Shanghai, China
| | - Zaiqi Wang
- InxMed (Shanghai) Co., Ltd., Shanghai, China
| |
Collapse
|
46
|
Zheng L, Cai W, Ke Y, Hu X, Yang C, Zhang R, Wu H, Liu D, Yu H, Wu C. Cancer‑associated fibroblasts: a pivotal regulator of tumor microenvironment in the context of radiotherapy. Cell Commun Signal 2025; 23:147. [PMID: 40114180 PMCID: PMC11927177 DOI: 10.1186/s12964-025-02138-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND In the course of tumor treatment, radiation therapy (RT) not only kills cancer cells, but also induces complex biological effects in non-malignant cells around cancer cells. These biological effects such as angiogenesis, changes in stromal composition and immune cell infiltration remodel the tumor microenvironment (TME). As one of the major components of the TME, Cancer‑associated fibroblasts (CAFs) are not only involved in tumorigenesis, progression, recurrence, and metastasis but also regulate the tumor-associated immune microenvironment. CAFs and tumor cells or immune cells have complex intercellular communication in the context of tumor radiation. MAIN CONTENT Different cellular precursors, spatial location differences, absence of specific markers, and advances in single-cell sequencing technology have gradually made the abundant heterogeneity of CAFs well known. Due to unique radioresistance properties, CAFs can survive under high doses of ionizing radiation. However, radiation can induce phenotypic and functional changes in CAFs and further act on tumor cells and immune cells to promote or inhibit tumor progression. To date, the effect of RT on CAFs and the effect of irradiated CAFs on tumor progression and TME are still not well defined. CONCLUSION In this review, we review the origin, phenotypic, and functional heterogeneity of CAFs and describe the effects of RT on CAFs, focusing on the mutual crosstalk between CAFs and tumor or immune cells after radiation. We also discuss emerging strategies for targeted CAFs therapy.
Collapse
Affiliation(s)
- Linhui Zheng
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Wenqi Cai
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Yuan Ke
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Xiaoyan Hu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Chunqian Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Runze Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Huachao Wu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Dong Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Haijun Yu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China.
- Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071, China.
| | - Chaoyan Wu
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China.
| |
Collapse
|
47
|
Kirkil G, Mogulkoc N, Jovanovic D. Risk factors and management of lung cancer in idiopathic pulmonary fibrosis: A comprehensive review. SARCOIDOSIS, VASCULITIS, AND DIFFUSE LUNG DISEASES : OFFICIAL JOURNAL OF WASOG 2025; 42:15604. [PMID: 40100103 PMCID: PMC12013682 DOI: 10.36141/svdld.v42i1.15604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 08/12/2024] [Indexed: 03/20/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease. Lung cancer (LC) is among the most crucial comorbidity factors in patients with IPF. IPF patients that are diagnosed with LC have a reduced mean survival time. Therapeutic strategies for LC in patients with IPF need to be adapted according to the individual treatment risk. Life-threatening acute exacerbation (AE) of IPF may occur in association with cancer treatment, thereby severely restricting the therapeutic options for IPF-associated LC. Because LC and anticancer treatments can worsen the prognosis of IPF, the prevention of LC is as critical as managing patients with IPF.
Collapse
Affiliation(s)
- Gamze Kirkil
- Department of Chest Disease, University of Firat, Elazig, Türkiye
| | - Nesrin Mogulkoc
- Department of Chest Disease, University of Ege, İzmir, Türkiye
| | | |
Collapse
|
48
|
Chen J, Wang Q, Wu H, Huang X, Cao C. Therapies targeting triple-negative breast cancer: a perspective on anti-FGFR. Front Oncol 2025; 14:1415820. [PMID: 40135140 PMCID: PMC11932845 DOI: 10.3389/fonc.2024.1415820] [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: 04/11/2024] [Accepted: 09/02/2024] [Indexed: 03/27/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the subtypes with the worst prognosis due to tumour heterogeneity and lack of appropriate treatment. This condition is a consequence of the distinctive tumour microenvironment (TME). The TME is associated with factors such as the promotion of proliferation, angiogenesis, inhibition of apoptosis, suppression of the immune system and drug resistance. Therefore, remodelling the TME is critical for the treatment of TNBC. A key role in the formation of the TME is played by the fibroblast growth factor/fibroblast growth factor receptor(FGF/FGFR) signalling pathway. Thus, the FGFRs may be a potential target for treating TNBC. Over-activated FGFRs promote growth, migration and drug resistance in TNBC by influencing the onset of TME events, tumour angiogenesis and immune rejection. A thorough comprehension of the FGF/FGFR signalling pathway's mechanism of action in the development of TNBC could offer valuable insights for discovering new therapeutic strategies and drug targets. Inhibiting the FGF/FGFR axis could potentially hinder the growth of TNBC and its drug resistance by disrupting crucial biological processes in the TME, such as angiogenesis and immune evasion. This review evaluates the potential of inhibiting the FGF/FGFR axis as a strategy for treating TNBC. It explores the prospects for developing related therapeutic approaches. This study explores the research and application prospects of the FGF/FGFR axis in TNBC. The aim is to provide guidance for further therapeutic research and facilitate the development of innovative approaches targeting TNBC.
Collapse
Affiliation(s)
- Jinhao Chen
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei, China
- Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei, China
- College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Qianru Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei, China
- College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Hongyan Wu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei, China
- College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Xiaofei Huang
- Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei, China
- College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Chunyu Cao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei, China
- College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| |
Collapse
|
49
|
Liao T, Chen X, Qiu F, Zhang X, Wu F, Zhao Z, Xu M, Chen M, Shen JW, Shen Q, Ji J. Regulation of cancer-associated fibroblasts for enhanced cancer immunotherapy using advanced functional nanomedicines: an updated review. J Nanobiotechnology 2025; 23:166. [PMID: 40038745 PMCID: PMC11877876 DOI: 10.1186/s12951-025-03217-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
The tumor microenvironment (TME) is a complex and dynamic ecosystem that plays a critical role in cancer progression. It comprises various cell types, including immune cells, tumor cells, and stromal cells. Among these, cancer-associated fibroblasts (CAFs) represent a heterogeneous population with diverse origins, phenotypes, and functions. Activated CAFs secrete multiple factors that promote tumor growth, migration, angiogenesis, and contribute to chemoresistance. Additionally, CAFs secrete extracellular matrix (ECM) components, such as collagen, which form a physical barrier that hinders the penetration of chemotherapeutic and immunotherapeutic agents. This ECM also influences immune cell infiltration, impeding their ability to effectively target tumor cells. As a result, modulating the activity of CAFs has emerged as a promising strategy to enhance the efficacy of tumor immunotherapy. Nano-delivery systems, constructed from various nanomaterials with high targeting specificity and biocompatibility, offer a compelling approach to deliver therapeutic agents or immunomodulatory factors directly to CAFs. This modulation can alter CAF function, reduce their tumor-promoting effects, and thereby improve the outcomes of immunotherapy. This review provides an in-depth exploration of the origins, functions, and interactions of CAFs within the TME, particularly in the context of immune suppression. Furthermore, it discusses the potential applications of functional nanocarrifers in modulating CAFs and enhancing the effectiveness of tumor immunotherapy, highlighting the significant progress and potential of nanotechnology in this area.
Collapse
Affiliation(s)
- Tingting Liao
- School of Pharmacy, College of Pharmacy, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, 310015, Zhejiang, China
| | - Xiaoxiao Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Fengkai Qiu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Xinyu Zhang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, China
| | - Fazong Wu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Zhongwei Zhao
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Ming Xu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Minjiang Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, China
| | - Jia-Wei Shen
- School of Pharmacy, College of Pharmacy, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, 310015, Zhejiang, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, China.
| | - Qiying Shen
- School of Pharmacy, College of Pharmacy, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, 310015, Zhejiang, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, China.
| | - Jiansong Ji
- School of Pharmacy, College of Pharmacy, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, 310015, Zhejiang, China.
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China.
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| |
Collapse
|
50
|
Meguro S, Nakanishi M. Cellular senescence in the cancer microenvironment. J Biochem 2025; 177:171-176. [PMID: 39760850 DOI: 10.1093/jb/mvaf001] [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: 09/29/2024] [Revised: 11/25/2024] [Accepted: 12/07/2024] [Indexed: 01/07/2025] Open
Abstract
In this ageing society, the number of patients suffering from age-related diseases, including cancer, is increasing. Cellular senescence is a cell fate that involves permanent cell cycle arrest. Accumulated senescent cells in tissues over time present senescence-associated secretory phenotype (SASP) and make the inflammatory context, disturbing the tumour microenvironment. In particular, the effect of senescent cancer-associated fibroblasts on cancer progression has recently come under the spotlight. Although scientific evidence on the impact of cellular senescence on cancer is emerging, the association between cellular senescence and cancer is heterogeneous and the comprehensive mechanism is still not revealed. Recently, a therapy targeting senescent cells, senotherapeutics, has been reported to be effective against cancer in preclinical research and even clinical trials. With further research, the development of senotherapeutics as a novel cancer therapy is expected.
Collapse
Affiliation(s)
- Satoru Meguro
- Division of Cancer Cell Biology, Institute of Medical Science, University of Tokyo, Shirokanedai 4-6-1, Minato-ku, Tokyo 108-8639, Tokyo, Japan
- Department of Urology, Fukushima Medical University School of Medicine, Fukushima, 960-1247, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, Institute of Medical Science, University of Tokyo, Shirokanedai 4-6-1, Minato-ku, Tokyo 108-8639, Tokyo, Japan
| |
Collapse
|