1
|
Zheng C, Hei H, Zhai Y, Gong W, Zhang R, Zhang S. CAFs-released exosomal CREB1 promotes cell progression and immune evasion in thyroid cancer via the positive regulation of CCL20. Autoimmunity 2025; 58:2458324. [PMID: 39863628 DOI: 10.1080/08916934.2025.2458324] [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: 11/12/2024] [Revised: 01/10/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Exosomes derived from cancer-associated fibroblasts (CAFs) can affect tumor microenvironment (TME) of thyroid cancer (TC). The cAMP response element binding protein 1 (CREB1) acts as a transcription factor to participate in cancer development. Currently, we aimed to explore the molecular mechanism of exosome-associated CREB1 and C-C motif chemokine ligand 20 (CCL20) in TC. METHODS The mRNA and protein levels were examined via RT-qPCR and western blot. Gene interaction was analyzed using ChIP and dual-luciferase reporter assays. Cell migration, invasion and proliferation were assessed by wound healing, transwell and EdU assays. Exosomes were characterized by morphology observation and western blot. The proliferation and apoptosis of CD8+ T cells were detected by immunofluorescence and flow cytometry. In vivo assays were performed by establishing xenograft models. RESULTS CREB1 was highly expressed in TC. CREB1 positively interacted with CCL20 in TC. CREB1 facilitated TC cell migration, invasion and proliferation via targeting CCL20. CCL20 expression was reduced by transferring CAFs-secreted exosomes sheltering CREB1 downregulation. Exosomal CREB1 knockdown receded cell progression and enhanced CD8+ T function by mediating CCL20. CAFs-associated exosomal CREB1 downregulation inhibited tumorigenesis through affecting CCL20. CONCLUSION CAFs-derived exosomes accelerated the malignant behaviors and immune evasion in TC by carrying CREB1 to up-regulate CCL20.
Collapse
Affiliation(s)
- Chen Zheng
- Department of Thyroid Head and Neck Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Hu Hei
- Department of Thyroid Head and Neck Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yifei Zhai
- Department of Thyroid Head and Neck Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Wenbo Gong
- Department of Thyroid Head and Neck Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Runfang Zhang
- Department of Thyroid Head and Neck Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Songtao Zhang
- Department of Thyroid Head and Neck Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
2
|
Chen T, Xu Y, Yang F, Pan Y, Ji N, Li J, Zeng X, Chen Q, Jiang L, Shen YQ. Crosstalk of glutamine metabolism between cancer-associated fibroblasts and cancer cells. Cell Signal 2025; 133:111874. [PMID: 40381975 DOI: 10.1016/j.cellsig.2025.111874] [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: 04/14/2025] [Revised: 05/06/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Glutamine (Gln), a critical metabolic substrate, fuels the uncontrolled proliferation of cancer cells. Cancer-associated fibroblasts (CAFs), essential components of the tumor microenvironment, facilitate tumor progression by supplying Gln to cancer cells and driving drug resistance through metabolic reprogramming. This review highlights the key processes of Gln uptake, transport, and catabolism and explores the metabolic crosstalk between CAFs and cancer cells. It also examines the roles of major oncogenic regulators-c-Myc, mTORC, KRAS, p53, and HIF-in controlling Gln metabolism and shaping therapeutic resistance. Current pharmacological approaches targeting Gln metabolism, including enzyme inhibitors and transporter blockers, are discussed alongside emerging therapeutic strategies and ongoing clinical trials. Lastly, we underscore the importance of integrating advanced technologies like artificial intelligence and spatial omics to refine treatment targeting and develop more effective, personalized therapeutic interventions.
Collapse
Affiliation(s)
- Tingyu Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yiming Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fan Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanxin Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ning Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lu Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ying-Qiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
3
|
Malhotra P, Fyfe J, Emmanouilidi A, Casari I, Mellett NA, Huynh K, Pajic M, Greening DW, Meikle PJ, Falasca M. Oncogenic small extracellular vesicles enriched in sphingosine-1-phosphate play a crucial role in pancreatic cancer progression. Cell Signal 2025; 132:111775. [PMID: 40158707 DOI: 10.1016/j.cellsig.2025.111775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Small extracellular vesicles (sEVs) from tumour cells mediate intercellular communication and signalling to regulate the progression of pancreatic ductal adenocarcinoma (PDAC). While we and others have shown that PDAC-derived sEVs comprise oncogenic protein and nucleic acid cargo, understanding the lipid landscape of these sEVs remains unknown. Lipids influence both the composition of sEVs and their roles in lipid metabolism and signalling pathways within the tumour microenvironment and tumorigenesis. We hypothesised that specific lipids in oncogenic sEVs might provide insights into PDAC. Comprehensive mass spectrometry-based lipidomic analysis was performed using liquid chromatography-electrospray ionisation-tandem mass spectrometry on sEVs isolated from PDAC and non-malignant pancreatic cell lines, patient-derived xenograft cell lines and plasma from the PDAC transgenic mouse model KPC (KRASWT/G12D/ TP53WT/R172H/Pdx1-Cre+/+). The sEV lipidomic analyses identified over 700 lipid species from 25 lipid classes and subclasses. Our results showed that, compared to non-malignant cells, PDAC-derived sEVs were enriched in specific lysophospholipids, particularly sphingosine-1-phosphate (S1P), a lipid known for its pivotal role in cancer pathogenesis. S1P enrichment was validated in plasma-derived sEVs from KPC mice compared to WT. To explore the functional implications of S1P enrichment, we conducted assays demonstrating that S1P in sEVs facilitated tubule formation in human microvascular endothelial cells and promoted cancer-associated fibroblast cell migration. We show that PDAC-derived sEVs are differentially enriched in specific lipids associated with cancer phenotype. Our findings highlight that PDAC-derived sEVs are enriched in specific lipids, particularly S1P, which plays a crucial role in promoting cancer progression.
Collapse
Affiliation(s)
- Pratibha Malhotra
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Jordan Fyfe
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Aikaterini Emmanouilidi
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Ilaria Casari
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Natalie A Mellett
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Kevin Huynh
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Marina Pajic
- Translational Oncology Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - David W Greening
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia; Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia; School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Marco Falasca
- University of Parma, Department of Medicine and Surgery, Via Volturno 39, 43125 Parma, Italy.
| |
Collapse
|
4
|
Sagliocchi S, Acampora L, Barone B, Crocetto F, Dentice M. The impact of the tumor microenvironment in the dual burden of obesity-cancer link. Semin Cancer Biol 2025; 112:36-42. [PMID: 40127706 DOI: 10.1016/j.semcancer.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 03/26/2025]
Abstract
Obesity induces systemic perturbations of tissue homeostasis, leading to dyslipidemia, insulin resistance and chronic state of inflammation. Evidence from clinical and preclinical studies links excess of adiposity with increased cancer incidence and suggests that chronic inflammation may contribute to increased cancer risk in obese patients. Over the last decades of obesity research, multifaced and complicated effects of abnormal or excessive expansion of Adipose Tissue have been uncovered. In particular, it is widely described how obesity can exacerbate the tumorigenesis for instance by fueling soluble signals and adipokines and by enhancing tissue inflammation and altering the hormonal balance. Less is known about the paracrine effects of the cancer-associated adipocytes on the tumor cells and still poorly explored is the reciprocal communication between cancer cells and the adipose component of the tumor microenvironment (TME). In this review, we will address the mechanisms by which the peritumoral Adipose Tissue can influence the dynamics of tumoral cells. We will discuss how obesity-induced changes in the tumor microenvironment may enhance tumor growth and aggressive characteristics leading to increased invasiveness and metastatic progression of cancer that leads to a worsen cancer survival in obese subjects. We conclude that targeting the peritumoral adipose component of the TME would be a therapeutic option to prevent cancer development.
Collapse
Affiliation(s)
- Serena Sagliocchi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Lucia Acampora
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Biagio Barone
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples 80131, Italy
| | - Felice Crocetto
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples 80131, Italy
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy; CEINGE - Biotecnologie Avanzate Scarl, Naples, Italy.
| |
Collapse
|
5
|
Cheng Z, Yang X, Ren Y, Wang H, Zhang Q, Lin S, Wu W, Yang X, Zheng J, Liu X, Tao X, Chen X, Qian Y, Li X. Investigating the molecular mechanisms and clinical potential of APO+ endothelial cells associated with PANoptosis in the tumor microenvironment of hepatocellular carcinoma using single-cell sequencing data. Transl Oncol 2025; 57:102402. [PMID: 40318262 PMCID: PMC12123355 DOI: 10.1016/j.tranon.2025.102402] [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: 02/24/2025] [Revised: 03/27/2025] [Accepted: 04/19/2025] [Indexed: 05/07/2025] Open
Abstract
INTRODUCTION PANoptosis is a newly identified form of programmed cell death that integrates elements of pyroptosis, apoptosis, and necroptosis. It plays a pivotal role in shaping the tumor immune microenvironment. Despite its significance, the specific functions and mechanisms of PANoptosis within the tumor microenvironment (TME) of hepatocellular carcinoma (HCC) remain unclear. This study aims to investigate these mechanisms using single-cell RNA sequencing data. METHODS Single-cell RNA sequencing data from HCC patients were obtained from the GEO database. The AUCell algorithm was used to quantify PANoptosis activity across various cell types in the TME. Cell populations with high PANoptosis scores were further analyzed using CytoTRACE and scMetabolism to assess their differentiation states and metabolic profiles. Associations between these high-score cell subsets and patient prognosis, tumor stage, and response to immunotherapy were examined. Cell-cell communication analysis was performed to explore how PANoptosis-related APO+ endothelial cells (ECs) may influence HCC progression. Immunofluorescence staining was used to assess the spatial distribution of APO+ ECs in tumor and adjacent tissues. Finally, a CCK8 assay was conducted to evaluate the effect of APOH+ HUVECs on HCC cell proliferation. RESULTS A total of 16 HCC patient samples with single-cell RNA sequencing data were included in the study. By calculating the PANoptosis scores of different cell types, we found that ECs, macrophages, hepatocytes, and fibroblasts exhibited higher PANoptosis scores. The PANoptosis scores, differentiation trajectories, intercellular communication, and metabolic characteristics of these four cell subpopulations with high PANoptosis scores were visualized. Among all subpopulations, APO+ ECs demonstrated the most significant clinical relevance, showing a positive correlation with better clinical staging, prognosis, and response to immunotherapy in HCC patients. Cellular communication analysis further revealed that APO+ ECs might regulate the expression of HLA molecules, thereby influencing T cell proliferation and differentiation, potentially contributing to improved prognosis in HCC patients. Immunofluorescence staining results indicated that APO+ ECs were primarily located in the adjacent tissues of HCC patients, with lower expression in tumor tissues. The results of cellular experiments showed that APOH+ HUVECs significantly inhibited the proliferation of HCC cells. CONCLUSIONS This study systematically mapped the cellular landscape of the TME in HCC patients and explored the differences in differentiation trajectories, metabolic pathways, and other aspects of subpopulations with high PANoptosis scores. Additionally, the study elucidated the potential molecular mechanisms through which APO+ ECs inhibit HCC cell proliferation and improve prognosis and immunotherapeutic efficacy in HCC patients. This research provides new insights for clinical prognosis evaluation and immunotherapy strategies in HCC.
Collapse
Affiliation(s)
- Zhaorui Cheng
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China; Department of Urology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Xiangyu Yang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, China.
| | - Yi Ren
- Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Huimin Wang
- Department of Traditional Chinese Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Qi Zhang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Sailing Lin
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Wenhao Wu
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Xiaolu Yang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Jiahan Zheng
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xinzhu Liu
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Xin Tao
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang, JiangXi, China
| | - Xiaoyong Chen
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China.
| | - Yuxin Qian
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China; Department of Urology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Xiushen Li
- Department of Traditional Chinese Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, China; Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China.
| |
Collapse
|
6
|
Huang Y, Chen L, Chen Y, Zhou S, Xie X, Xie J, Yu M, Chen J. High-density lipoprotein-based nanoplatform reprograms tumor microenvironment and enhances chemotherapy against pancreatic ductal adenocarcinoma. Biomaterials 2025; 318:123147. [PMID: 39908877 DOI: 10.1016/j.biomaterials.2025.123147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/25/2024] [Accepted: 01/26/2025] [Indexed: 02/07/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly aggressive, with limited success in traditional therapies due to the fibrotic, immunosuppressive, pro-metastatic tumor microenvironment (TME), which collectively impede the drug accumulation and accelerate the tumor progression. In this work, we developed a PDAC-customized nutrient-mimicking reconstituted high-density lipoprotein (rHDL) capable of efficiently co-encapsulate versatile TME regulating cannabidiol and cytotoxic gemcitabine to simultaneously reprogram TME while suppressing PDAC progression. Specifically, a small-sized, nutrient-like rHDL was constructed to realize deep PDAC parenchyma penetration and efficient intra-tumoral uptake. Next, natural herbal compound cannabidiol was screened and incorporated into rHDL to regulate TME via attenuating fibrosis, reliving immunosuppression and mitigating metastatic tendency. At last, gemcitabine, the PDAC gold standard first-line therapy was co-delivered by the PDAC-customized rHDL to overcome drug resistance and amplify its PDAC suppression. Our findings demonstrate the feasibility of an integrated multi-stage TME regulation strategy for improved PDAC therapy, and might represent a modality in promoting chemotherapy against PDAC.
Collapse
Affiliation(s)
- Yukun Huang
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China; Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liang Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Yu Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Songlei Zhou
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Xiaoying Xie
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Jing Xie
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Minghua Yu
- Fudan University Clinical Research Center for Cell-based Immunotherapy & Department of Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Shanghai, 201399, China
| | - Jun Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
7
|
Gerakopoulos V, Ramos C, Müller C, Walterskirchen N, Vintila S, Zotter C, Ilg M, Pap A, Riss S, Bergmann M, Unger LW, Vogt AB, Oehler R, Lukowski SW. Single-cell transcriptomic analysis identifies tissue-specific fibroblasts as the main modulators of myeloid cells in peritoneal metastasis of different origin. Cancer Lett 2025; 620:217678. [PMID: 40154914 DOI: 10.1016/j.canlet.2025.217678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/12/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Colorectal cancer (CRC) peritoneal metastasis (CPM) is related to limited therapy options and poor prognosis. Although stromal cells heavily infiltrate most CPMs, interactions between different cell types in their microenvironment remain unclear. Here, we investigated tumor and distant normal tissue from CPM and CRC patients using single-cell RNA sequencing. Investigating the incoming and outgoing signals between cells revealed that fibroblasts dominate the CPM signaling landscape with myeloid cells as their strongest interaction partner. Using immunohistochemistry, we confirmed that fibroblasts co-localize with macrophages in the CPM microenvironment. A fibroblast sub-population detected only in CPM and normal peritoneum demonstrated immunoregulatory properties in co-culture experiments, and was further detected in additional peritoneal malignancies derived from ovarian and gastric origin. This novel fibroblast type and its communication with macrophages could be attractive targets for therapeutic interventions in CPM and potentially peritoneal surface malignancies in general.
Collapse
Affiliation(s)
- Vasileios Gerakopoulos
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Cristiano Ramos
- 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
| | - Natalie Walterskirchen
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Stefania Vintila
- Division of Visceral 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
| | - Mathias Ilg
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120, Vienna, Austria
| | - Anna Pap
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120, Vienna, Austria
| | - Stefan Riss
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael Bergmann
- Division of Visceral 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; Dept. of Colorectal Surgery, Oxford University Hospitals, Old Rd, Headington, Oxford, OX3 7LE, United Kingdom
| | - Anne B Vogt
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120, Vienna, Austria
| | - Rudolf Oehler
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria.
| | - Samuel W Lukowski
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120, Vienna, Austria
| |
Collapse
|
8
|
Issa H, Singh L, Lai KS, Parusheva-Borsitzky T, Ansari S. Dynamics of inflammatory signals within the tumor microenvironment. World J Exp Med 2025; 15:102285. [DOI: 10.5493/wjem.v15.i2.102285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/31/2024] [Accepted: 01/11/2025] [Indexed: 04/16/2025] Open
Abstract
Tumor stroma, or tumor microenvironment (TME), has been in the spotlight during recent years for its role in tumor development, growth, and metastasis. It consists of a myriad of elements, including tumor-associated macrophages, cancer-associated fibroblasts, a deregulated extracellular matrix, endothelial cells, and vascular vessels. The release of proinflammatory molecules, due to the inflamed microenvironment, such as cytokines and chemokines is found to play a pivotal role in progression of cancer and response to therapy. This review discusses the major key players and important chemical inflammatory signals released in the TME. Furthermore, the latest breakthroughs in cytokine-mediated crosstalk between immune cells and cancer cells have been highlighted. In addition, recent updates on alterations in cytokine signaling between chronic inflammation and malignant TME have also been reviewed.
Collapse
Affiliation(s)
- Hala Issa
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Lokjan Singh
- Department of Microbiology, Karnali Academy of Health Sciences, Jumla 21200, Karnali, Nepal
| | - Kok-Song Lai
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Tina Parusheva-Borsitzky
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Shamshul Ansari
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| |
Collapse
|
9
|
Kim M, Choi R, Kim L, Kim YC, Noh I. Cell membrane nanoparticles in cancer therapy: From basic structure to surface functionalization. J Control Release 2025; 382:113752. [PMID: 40254140 DOI: 10.1016/j.jconrel.2025.113752] [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: 02/21/2025] [Revised: 04/06/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Cell membrane nanoparticles (CNPs) have recently garnered significant attention as effective drug-delivery vehicles. Beyond their simple function of encapsulating cargo within a lipid bilayer structure, the cell membrane is a complex entity derived from biological materials, presenting a variety of surface proteins and glycans. Notable features that enhance their effectiveness as delivery vehicles include the inhibition of protein corona formation in the plasma and the suppression of macrophage phagocytosis, both of which contribute to prolonged blood circulation. Furthermore, CNPs exhibit homotypic targeting effects toward their cells of origin, resulting in reduced side effects, and because they are not xenobiotics, the likelihood of nonspecific immune activation is also minimized. This review focuses on various applications of CNPs in cancer therapeutic studies, examining their structural evolution and surface engineering developments. We introduce studies that leverage the inherent functionality of cell membranes and recent research in functional CNPs synthesized through genetic or chemical engineering methods. Through this review, we aim to trace the progression of CNP research, explore potential directions for their use in biomedical applications, and assess the prospects for clinical trials.
Collapse
Affiliation(s)
- Munsik Kim
- Department of Medical Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea; Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea; Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Rohbin Choi
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Lian Kim
- Department of Medical Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yeu-Chun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| | - Ilkoo Noh
- Department of Medical Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea; Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea.
| |
Collapse
|
10
|
Gong L, Wu L, Zhao S, Xiao S, Chu X, Zhang Y, Li F, Li S, Yang H, Jiang P. Epigenetic regulation of ferroptosis in gastrointestinal cancers (Review). Int J Mol Med 2025; 55:93. [PMID: 40242977 PMCID: PMC12045471 DOI: 10.3892/ijmm.2025.5534] [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: 11/13/2024] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Ferroptosis is a type of iron‑dependent cell death characterized by excessive lipid peroxidation and may serve as a potential therapeutic target in cancer treatment. While the mechanisms governing ferroptosis continue to be explored and elucidated, an increasing body of research highlights the significant impact of epigenetic modifications on the sensitivity of cancer cells to ferroptosis. Epigenetic processes, such as DNA methylation, histone modifications and non‑coding RNAs, have been identified as key regulators that modulate the expression of ferroptosis‑related genes. These alterations can either enhance or inhibit the sensitivity of gastrointestinal cancer (GIC) cells to ferroptosis, thereby affecting the fate of GICs. Drugs that target epigenetic markers for advanced‑stage cancer have shown promising results in enhancing ferroptosis and inhibiting tumor growth. This review explores the intricate relationship between epigenetic regulation and ferroptosis in GICs. Additionally, the potential of leveraging epigenetic modifications to trigger ferroptosis in GICs is investigated. This review highlights the importance of further research to elucidate the specific mechanisms underlying epigenetic control of ferroptosis and to advance the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Linqiang Gong
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Linlin Wu
- Oncology Department, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shiyuan Zhao
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, Shandong 272000, P.R. China
| | - Shuai Xiao
- Department of Intensive Care Medicine, Tengzhou Central People's Hospital, Jining Medical University, Tengzhou, Shandong 277500, P.R. China
| | - Xue Chu
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
| | - Yazhou Zhang
- Department of Foot and Ankle Surgery, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Fengfeng Li
- Neurosurgery Department, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shuhui Li
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Hui Yang
- Department of Gynecology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, Shandong 272000, P.R. China
| |
Collapse
|
11
|
Collinson R, Tanos B. Primary cilia and cancer: a tale of many faces. Oncogene 2025; 44:1551-1566. [PMID: 40301543 PMCID: PMC12095056 DOI: 10.1038/s41388-025-03416-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/04/2025] [Accepted: 04/10/2025] [Indexed: 05/01/2025]
Abstract
Cilia are microtubule-based sensory organelles which project from the cell surface, enabling detection of mechanical and chemical stimuli from the extracellular environment. It has been shown that cilia are lost in some cancers, while others depend on cilia or ciliary signaling. Several oncogenic molecules, including tyrosine kinases, G-protein coupled receptors, cytosolic kinases, and their downstream effectors localize to cilia. The Hedgehog pathway, one of the most studied ciliary-signaling pathways, is regulated at the cilium via an interplay between Smoothened (an oncogene) and Patched (a tumor suppressor), resulting in the activation of pro-survival programs. Interestingly, cilia loss can result in resistance to Smoothened-targeting drugs and increased cancer cell survival. On the other hand, kinase inhibitor-resistant and chemoresistant cancers have increased cilia and increased Hedgehog pathway activation, and suppressing cilia can overcome this resistance. How cilia regulate cancer is therefore context dependent. Defining the signaling output of cilia-localized oncogenic pathways could identify specific targets for cancer therapy, including the cilium itself. Increasing evidence implicates cilia in supporting several hallmarks of cancer, including migration, invasion, and metabolic rewiring. While cell cycle cues regulate the biogenesis of cilia, the absence of cilia has not been conclusively shown to affect the cell cycle. Thus, a complex interplay between molecular signals, phosphorylation events and spatial regulation renders this fascinating organelle an important new player in cancer through roles that we are only starting to uncover. In this review, we discuss recent advances in our understanding of cilia as signaling platforms in cancer and the influence this plays in tumor development.
Collapse
Affiliation(s)
- Rebecca Collinson
- Centre for Genome Engineering and Maintenance, Department of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, London, UK
| | - Barbara Tanos
- Centre for Genome Engineering and Maintenance, Department of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, London, UK.
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Xiao Z, Puré E. The fibroinflammatory response in cancer. Nat Rev Cancer 2025; 25:399-425. [PMID: 40097577 DOI: 10.1038/s41568-025-00798-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/06/2025] [Indexed: 03/19/2025]
Abstract
Fibroinflammation refers to the highly integrated fibrogenic and inflammatory responses mediated by the concerted function of fibroblasts and innate immune cells in response to tissue perturbation. This process underlies the desmoplastic remodelling of the tumour microenvironment and thus plays an important role in tumour initiation, growth and metastasis. More specifically, fibroinflammation alters the biochemical and biomechanical signalling in malignant cells to promote their proliferation and survival and further supports an immunosuppressive microenvironment by polarizing the immune status of tumours. Additionally, the presence of fibroinflammation is often associated with therapeutic resistance. As such, there is increasing interest in targeting this process to normalize the tumour microenvironment and thus enhance the treatment of solid tumours. Herein, we review advances made in unravelling the complexity of cancer-associated fibroinflammation that can inform the rational design of therapies targeting this.
Collapse
Affiliation(s)
- Zebin Xiao
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Heiskanen L, Nissinen L, Siljamäki E, Knuutila JS, Pellinen T, Kallajoki M, Heino J, Riihilä P, Kähäri VM. C5aR1 Promotes Invasion, Metastasis, and Poor Prognosis in Cutaneous Squamous Cell Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:1158-1171. [PMID: 40056975 DOI: 10.1016/j.ajpath.2025.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 03/18/2025]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the most common metastatic skin cancer, and the metastatic from is associated with a poor prognosis. Here, the role of the complement C5a receptor C5aR1 was examined in the progression and metastasis of cSCC. C5aR1 expression was increased in cSCC cells in a three-dimensional spheroid coculture model in the presence of fibroblasts, and treatment with recombinant C5a enhanced the invasion of cSCC cells. Staining for C5aR1 was detected on the surface of tumor cells at the invasive edge of human cSCC xenografts in vivo. Metastatic and non-metastatic primary human cSCCs, premalignant and benign epidermal lesions, and normal skin for C5aR1 were stained with multiplex immunofluorescence and chromogenic immunohistochemistry. Increased expression of C5aR1 was observed on the surface of tumor cells and fibroblasts in invasive cSCCs and recessive dystrophic epidermolysis bullosa-associated cSCCs compared with cSCC in situ, actinic keratoses, seborrheic keratoses, and normal skin. Increased expression of C5aR1 on the tumor cell surface and in fibroblasts was associated with metastatic risk and poor disease-specific survival of patients with primary cSCC. These findings suggest a role of C5a in cSCC cell invasion, and they identify C5aR1 as a novel biomarker for metastasis risk and poor prognosis in patients with cSCC. The results also suggest that C5aR1 could be a novel therapeutic target for the treatment of locally advanced and metastatic cSCC.
Collapse
Affiliation(s)
- Lauri Heiskanen
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Liisa Nissinen
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Elina Siljamäki
- Department of Life Technologies and InFLAMES Research Flagship, University of Turku, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Jaakko S Knuutila
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Markku Kallajoki
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland
| | - Jyrki Heino
- Department of Life Technologies and InFLAMES Research Flagship, University of Turku, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Pilvi Riihilä
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Veli-Matti Kähäri
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland; FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland.
| |
Collapse
|
16
|
Hsu CY, Saleh RO, Mohammed JS, Mansuri N, Rekha MM, Kundlas M, Anand A, Sahoo S, Zwamel AH, Hulail HM. The dynamic interplay between melanoma cells and CAFs: Implications drug resistance and immune evasion and possible therapeutics. Exp Cell Res 2025; 449:114581. [PMID: 40311910 DOI: 10.1016/j.yexcr.2025.114581] [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: 03/13/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
Melanoma, a malignancy of varying prognoses across primary sites (cutaneous, ocular, and mucosal), typically displays peculiar treatment challenges in metastatic and refractory settings. Cancer-associated fibroblasts (CAFs) have long been recognized as pivotal components within melanoma's tumor microenvironment (TME), originating from various sources and manifesting considerable heterogeneity. These cells actively produce extracellular matrix (ECM), induce angiogenesis, provide metabolic support, contribute to drug resistance, and feed tumor progression and metastasis. Among the many growth factors and cytokines they secrete, including TGF-β and IL-6, they aid in anti-tumor immunity by recruiting immunosuppressive cells and inhibiting cytotoxic T-cell activity, contributing to immune evasion. These dynamic cells sculpt the tumor's niche, allowing cancer cells to survive and proliferate, and their existence is widely correlated with poor prognosis. Taking a cue from the previously established groundwork of how the surroundings heavily influence tumor development, this review attempts to profile the intricate interaction of melanoma cells with the CAFs, the ECM, and signaling molecules. We explore different subtypes of CAFs, their origins, and how they have evolved in their pro-tumorigenic roles in melanoma. Additionally, we review recent advancements in the therapeutic arsenal targeting CAFs to achieve a more effective treatment response. By detailing the specific roles played by different CAFs subtypes in the modulation of immuno-responses and treatment outcomes, this review will further provide insights into the targeted therapy to disrupt CAFs-mediated tumor support in melanoma.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University, Tempe Campus, Phoenix, AZ, 85004, USA.
| | - Raed Obaid Saleh
- Department of Medical Laboratories Techniques, College of Health and Medical Techniques, University of Al Maarif, Al Anbar, 31001, Iraq.
| | - Jaafaru Sani Mohammed
- Medical Analysis Department, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Nasrin Mansuri
- Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mayank Kundlas
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Alex Anand
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Samir Sahoo
- Department of General Medicine, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Ahmed Hussein Zwamel
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq; Department of medical analysis, Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Department of medical analysis, Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| | - Hanen Mahmod Hulail
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
17
|
Monteiro MV, Moreira-Silva F, Lagarto M, Ferreira LP, Ramalhinho C, Duarte IF, Jerónimo C, Gaspar VM, Mano JF. Bioengineered Tumor-Stroma Prostate Cancer In Vitro Models for Screening Therapeutics. Biotechnol Bioeng 2025; 122:1541-1553. [PMID: 40083131 DOI: 10.1002/bit.28971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/17/2025] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
Cancer-associated fibroblasts are increasingly recognized to have a high impact on prostate tumor growth and drug resistance. Here, we bioengineered organotypic prostate cancer 3D in vitro models to better understand tumor-stroma interplay, the metabolomic profile underlying such interactions, and their impact on standard-of-care therapeutics performance. The assembly of robust and uniform spheroids was evaluated and compared in monotypic PC-3 and heterotypic microtumors comprised of either a healthy or malignant stroma and prostate cancer cells. Our findings demonstrate that the precise inclusion of prostate cancer stromal elements is crucial to generating robust PC-3 prostate cancer spheroids with reproducible morphology and size. The inclusion of cancer-associated fibroblasts promoted the establishment of more compact microtumors exhibiting characteristic expression of major proteins. Exometabolomic profile analysis also highlighted the impact of stromal cells on tumor models metabolism. The optimized heterotypic spheroids were additionally exploited for screening standard-of-care therapeutics, exhibiting a higher resistance when compared to their monotypic counterparts. Our findings demonstrate that including stromal elements in PC-3 prostate cancer models is crucial for their use as increasingly organotypic testing platforms, being relevant for screening candidate anti-cancer therapeutics and for the discovery of potential combinations with emerging anti-stroma therapies.
Collapse
Affiliation(s)
- Maria V Monteiro
- Department of Chemistry, CICECO-Aveiro Institute of Materials/LAQV-REQUIMTE, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Filipa Moreira-Silva
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), Rua Dr. António Bernardino de Almeida, Porto, Portugal
| | - Matilde Lagarto
- Department of Chemistry, CICECO-Aveiro Institute of Materials/LAQV-REQUIMTE, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Luís P Ferreira
- Department of Chemistry, CICECO-Aveiro Institute of Materials/LAQV-REQUIMTE, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Carlota Ramalhinho
- Department of Chemistry, CICECO-Aveiro Institute of Materials/LAQV-REQUIMTE, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Iola F Duarte
- Department of Chemistry, CICECO-Aveiro Institute of Materials/LAQV-REQUIMTE, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), Rua Dr. António Bernardino de Almeida, Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS-School of Medicine and Biomedical Sciences-University of Porto, Porto, Portugal
| | - Vítor M Gaspar
- Department of Chemistry, CICECO-Aveiro Institute of Materials/LAQV-REQUIMTE, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO-Aveiro Institute of Materials/LAQV-REQUIMTE, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| |
Collapse
|
18
|
Kang M, Min C, Devarasou S, Shin JH. Classification of differentially activated groups of fibroblasts using morphodynamic and motile features. APL Bioeng 2025; 9:026116. [PMID: 40385989 PMCID: PMC12084086 DOI: 10.1063/5.0250502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 05/05/2025] [Indexed: 05/20/2025] Open
Abstract
Fibroblasts play essential roles in cancer progression, exhibiting activation states that can either promote or inhibit tumor growth. Understanding these differential activation states is critical for targeting the tumor microenvironment (TME) in cancer therapy. However, traditional molecular markers used to identify cancer-associated fibroblasts are limited by their co-expression across multiple fibroblast subtypes, making it difficult to distinguish specific activation states. Morphological and motility characteristics of fibroblasts reflect their underlying gene expression patterns and activation states, making these features valuable descriptors of fibroblast behavior. This study proposes an artificial intelligence-based classification framework to identify and characterize differentially activated fibroblasts by analyzing their morphodynamic and motile features. We extract these features from label-free live-cell imaging data of fibroblasts co-cultured with breast cancer cell lines using deep learning and machine learning algorithms. Our findings show that morphodynamic and motile features offer robust insights into fibroblast activation states, complementing molecular markers and overcoming their limitations. This biophysical state-based cellular classification framework provides a novel, comprehensive approach for characterizing fibroblast activation, with significant potential for advancing our understanding of the TME and informing targeted cancer therapies.
Collapse
Affiliation(s)
- Minwoo Kang
- Department of Mechanical Engineering, KAIST, 291 Daehak-Ro, Yuseong-Gu, Daejeon 34141, Republic of Korea
| | - Chanhong Min
- Department of Mechanical Engineering, KAIST, 291 Daehak-Ro, Yuseong-Gu, Daejeon 34141, Republic of Korea
| | - Somayadineshraj Devarasou
- Department of Mechanical Engineering, KAIST, 291 Daehak-Ro, Yuseong-Gu, Daejeon 34141, Republic of Korea
| | - Jennifer H. Shin
- Author to whom correspondence should be addressed:. Tel.: +82 42 350 3232
| |
Collapse
|
19
|
Li X, Zhao H, Jiang E, Liu P, Chen Y, Wang Y, Li J, Wu Y, Liu Z, Shang Z. ITGB1/FERMT1 mechanoactivation enhances CD44 characteristic stemness in oral squamous cell carcinoma via ubiquitin-dependent CK1α degradation. Oncogene 2025; 44:1530-1544. [PMID: 40044983 DOI: 10.1038/s41388-025-03317-z] [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: 08/20/2024] [Revised: 01/30/2025] [Accepted: 02/18/2025] [Indexed: 05/15/2025]
Abstract
Cancer stem cells (CSCs) contribute to chemotherapy resistance and poor prognosis, posing significant challenges in the treatment of oral squamous cell carcinoma. The extracellular matrix (ECM)-constructed microenvironment remodels the niche of CSCs. Yet mechanisms by which biophysical properties of ECM relate to CSCs remain undefined. Here, our findings link ECM mechanical stimuli to CSCs phenotype transition, and propose that ECM stiffening mechanoactivates tumor cells to dedifferentiate and acquire CD44+ stem cell-like characteristics through noncanonical mechanotransduction. ITGB1 senses and transduces biomechanical signals, while FERMT1 acts as an intracellular mechanotransduction downstream, activating CSCs. Mechanistically, FERMT1 promotes the proteasomal degradation of CK1α by E3 ubiquitin ligase MIB1, thereby triggering Wnt signaling pathway. Combining targeted ECM softening with mechanotransduction inhibition strategy significantly attenuates tumor stemness and chemoresistance in vivo. Therefore, our findings highlight the role of ECM in regulating CSCs via biomechanical-dependent manner, suggesting the ECM/ITGB1/FERMT1/Wnt axis as a promising therapeutic target for CSCs therapy.
Collapse
Affiliation(s)
- Xiang Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hui Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Erhui Jiang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Pan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yue Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ji Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yufei Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhenan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
20
|
Jeong JH, Shin D, Kim SY, Bae DJ, Sung YH, Koh EY, Kim J, Kim CJ, Park JS, Choi JK, Kim SC, Jun E. Spatial distribution and activation changes of T cells in pancreatic tumors according to KRAS mutation subtype. Cancer Lett 2025; 618:217641. [PMID: 40090570 DOI: 10.1016/j.canlet.2025.217641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/03/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
To enhance immunotherapy efficacy in pancreatic cancer, it is crucial to characterize its immune landscape and identify key factors driving immune alterations. To achieve this, we quantitatively analyzed the immune microenvironment using multiplex immunohistochemistry, assessing the spatial relationships between immune and tumor cells to correlate with patient survival rates and oncological factors. Additionally, through Whole Exome Sequencing analysis based on public data, we explored genetic mutations that could drive these compositions. Finally, we validated T cell (Tc) migration mechanisms using patient-derived tumor organoids with induced KRAS mutation subtypes. Through this approach, we obtained the following meaningful results. First, immune cells in pancreatic cancer are denser in stromal regions than near tumor cells, with higher Tc distribution linked to increased patient survival rates. Second, the distance between tumor and Tc was within 100 μm, with higher Tc density found within 15-30 μm of the tumor cells. Third, while increasing CAF levels correspond to higher Tc density, higher ECM density tends to decrease Tc presence. Fourth, compared to KRAS G12D, KRAS G12V mutation increases various immune cells, notably Tc, which is closely linked to a dramatic rise in vascular cells. Finally, Tc migration was enhanced in tumor organoids with the G12V mutation, attributed to a reduction in the secretion of immunosuppressive cytokines. Our results indicate that KRAS mutation subtypes influence immune cell composition and function in the pancreatic cancer microenvironment, leading to varied immunotherapy responses. This underscores the need for personalized immune therapeutics and research models specific to KRAS mutations.
Collapse
Affiliation(s)
- Ji Hye Jeong
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Dakyum Shin
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation Surgery, Department of General Surgery, Chosun University Hospital, 365, Pilmun-daero, Dong-gu, Gwangju Metropolitan City, 61453, Republic of Korea
| | - Sang-Yeob Kim
- Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Dong-Jun Bae
- PrismCDX, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Young Hoon Sung
- Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Cell and Genetic Engineering, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Eun-Young Koh
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jinju Kim
- Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Chong Jai Kim
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jae Soon Park
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea; SCL-KAIST Institute of Translational Research, Daejeon, 34141, Republic of Korea
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea; SCL-KAIST Institute of Translational Research, Daejeon, 34141, Republic of Korea.
| | - Song Cheol Kim
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Surgery, BK21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| | - Eunsung Jun
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul, 05505, Republic of Korea; Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea.
| |
Collapse
|
21
|
Zhang Y, Zuo A, Ba Y, Liu S, Chen J, Yang S, Weng S, Chen Y, Xu H, Luo P, Cheng Q, Tang B, Liu B, Zhang C, Yang J, Han X, Liu Z. Cancer-associated fibroblast-derived SEMA3C facilitates colorectal cancer liver metastasis via NRP2-mediated MAPK activation. Proc Natl Acad Sci U S A 2025; 122:e2423077122. [PMID: 40402249 DOI: 10.1073/pnas.2423077122] [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: 11/06/2024] [Accepted: 04/09/2025] [Indexed: 05/23/2025] Open
Abstract
Liver metastasis remains the predominant cause of mortality in patients with colorectal cancer (CRC). Nevertheless, the mechanisms underlying the initiation of colorectal cancer liver metastasis remain poorly elucidated. During the metastatic process of CRC cells from the primary site to the liver, we performed time-resolved analyses and identified a subset of tumor cells spatially located in the primary tumor and temporally distributed in the early stages of liver metastasis. These cells were termed liver metastasis-initiating cells (LMICs). LMICs exhibit high stemness, low proliferation, active interaction with surrounding stromal components, and a close association with liver metastasis. Notably, we found significant interactions between cancer-associated fibroblasts (CAFs) and LMICs via the SEMA3C-NRP2 receptor-ligand pair. Further in vivo and in vitro experiments confirmed that CAF-secreted SEMA3C could bind to the NRP2 receptor, which activates the MAPK pathway and promotes colorectal cancer liver metastasis. Our findings suggest potential therapeutic strategies for the early prevention of colorectal cancer liver metastasis.
Collapse
Affiliation(s)
- Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou 450052, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou 450052, Henan, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jingqi Chen
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yukang Chen
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410000, China
| | - Bufu Tang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Benyu Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Chuhan Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jingkuan Yang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou 450052, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou 450052, Henan, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou 450052, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou 450052, Henan, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
22
|
Domonkos L, Yusenko M, Kovacs G, Banyai D. Partial regression of conventional renal cell carcinoma displays markers of wound repair. J Clin Pathol 2025; 78:404-408. [PMID: 39433307 DOI: 10.1136/jcp-2024-209459] [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: 02/02/2024] [Accepted: 06/05/2024] [Indexed: 10/23/2024]
Abstract
AIMS During detailed analysis of H&E-stained histological slides of 710 unbiased conventional renal cell carcinomas (cRCCs), 141 tumours displayed partial regressive changes showing strong similarity to that of wound healing. We aimed to analyse the molecular processes occurring in regressive tumours. METHODS Immunohistochemistry was applied to analyse the signalling molecules in 12 selected tumours, and statistical analysis was used to estimate the correlation between regression and the outcome of the disease. RESULTS The regressive areas displayed inflammatory granulation tissue expressing transforming growth factor beta-1 (TGFB1), interleukin-1 beta and interleukin-6 (IL1B and IL6), proliferation of alpha smooth muscle actin (αSMA) positive naïve activated fibroblasts and a diffuse fibronectin 1 (FN1) network. In the central areas of regressive tissues, parallel-running myofibroblasts showed FN1, collagen type I alpha 1 (COL1A1) and collagen type III alpha 1 (COL3A1) positive immunoreaction. Partial tumour regression is associated with a better postoperative course of the disease. CONCLUSIONS Partial regression is a frequent event in cRCCs. Recognising complex molecular processes involved in tumour regression might help to find a way towards 'healing' cRCC.
Collapse
Affiliation(s)
| | - Maria Yusenko
- Ruhr University Bochum, Bochum, Nordrhein-Westfalen, Germany
| | - Gyula Kovacs
- University of Pecs Medical School, Pecs, Hungary
| | | |
Collapse
|
23
|
Zhu X, Zhang L, Yu X, Yan P, Zhang X, Zhao Y, Wang D, Yang XA. Elucidating the tumor microenvironment interactions in breast, cervical, and ovarian cancer through single-cell RNA sequencing. Sci Rep 2025; 15:17846. [PMID: 40404741 PMCID: PMC12098903 DOI: 10.1038/s41598-025-03017-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 05/19/2025] [Indexed: 05/24/2025] Open
Abstract
This study aimed to identify the key cell types and their interactions in gynecological oncology of breast cancer, cervical cancer, and ovarian cancer. Single-cell RNA sequencing was performed on tumor samples of gynecological oncology from the GEO database. Cell types were identified using SingleR and cell composition was analyzed to understand the tumor microenvironment (TME). CellChat was used to analyze cell interactions, and pseudotemporal analysis was conducted on cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs) to understand their differentiation status. Four CAF subtypes were identified: iCAF, myCAF, proCAF, and matCAF. The iCAF subpopulation secreted COL1A1 and promoted tumor cell migration, while myCAF was involved in angiogenesis. The matCAF subpopulation was present throughout tumor development. TAMs were found to promote angiogenesis through the VEGFA_VEGFR2 signaling pathway. CAFs and TAMs play pivotal roles in tumor progression through their interactions and signaling pathways.
Collapse
Affiliation(s)
- Xiaoyue Zhu
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Liang Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Cardiology, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Xiaomin Yu
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
| | - Pengxian Yan
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
| | - Xiaoyu Zhang
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Yunlong Zhao
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Dongze Wang
- Clinical and Basic Medical College, Shandong First Medical University, Jinan, 250000, China
| | - Xiu-An Yang
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Anyuan Road, Chengde, 067000, China.
- Hebei Key Laboratory of Nerve Injury and Repair, Chengde Medical University, Chengde, 067000, China.
| |
Collapse
|
24
|
Bandyopadhyay A, Sinha S, Roy R, Biswas N. Autophagy mediated immune response regulation and drug resistance in cancer. Mol Biol Rep 2025; 52:492. [PMID: 40402380 DOI: 10.1007/s11033-025-10573-5] [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: 02/14/2025] [Accepted: 05/02/2025] [Indexed: 05/23/2025]
Abstract
Autophagy is a critical regulator of cellular homeostasis. The proteins involved in autophagy orchestrate the functions to strike the balance between cell survival and cell death in context-specific situations like aging, infections, inflammation and most importantly carcinogenesis. One of the major dead-locks in cancer treatment is the development of resistance to the available drugs (multi-drug resistance) as well as immune-suppressions in patients. Different studies over time have shown that autophagy is being involved in chemotherapy by working hand in hand with apoptosis or drug resistance through proliferative signals. Resistance to various drugs, such as, Cisplatin, Vincristine, Tamoxifen (TAM) occurs by epigenetic modifications, changed expression levels of microRNAs (miRNAs/miRs), and long non-coding RNAs (lncRNAs), which are regulated by the aberrant autophagy levels in lung, and breast cancers. More interestingly in the tumour microenvironment the immune suppressor cells also bring in autophagy in different pathway regulations either helping or opposing the whole carcinogenesis process. Macrophages, T cells, B cells, dendritic cells (DCs), neutrophils, and fibroblasts show involvement of autophagy in their differentiation and development in the tumor microenvironment (TME). Here, this extensive review for the first time tries to bring under a single canopy, several recent examples of autophagy-mediated immune regulations and autophagy-mediated epigenetically regulated drug resistance in different types of cancers.
Collapse
Affiliation(s)
- Anupriya Bandyopadhyay
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Samraj Sinha
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Rajdeep Roy
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Nabendu Biswas
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India.
| |
Collapse
|
25
|
Pentimalli TM, Schallenberg S, León-Periñán D, Legnini I, Theurillat I, Thomas G, Boltengagen A, Fritzsche S, Nimo J, Ruff L, Dernbach G, Jurmeister P, Murphy S, Gregory MT, Liang Y, Cordenonsi M, Piccolo S, Coscia F, Woehler A, Karaiskos N, Klauschen F, Rajewsky N. Combining spatial transcriptomics and ECM imaging in 3D for mapping cellular interactions in the tumor microenvironment. Cell Syst 2025; 16:101261. [PMID: 40220761 DOI: 10.1016/j.cels.2025.101261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/13/2024] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
Tumors are complex ecosystems composed of malignant and non-malignant cells embedded in a dynamic extracellular matrix (ECM). In the tumor microenvironment, molecular phenotypes are controlled by cell-cell and ECM interactions in 3D cellular neighborhoods (CNs). While their inhibition can impede tumor progression, routine molecular tumor profiling fails to capture cellular interactions. Single-cell spatial transcriptomics (ST) maps receptor-ligand interactions but usually remains limited to 2D tissue sections and lacks ECM readouts. Here, we integrate 3D ST with ECM imaging in serial sections from one clinical lung carcinoma to systematically quantify molecular states, cell-cell interactions, and ECM remodeling in CN. Our integrative analysis pinpointed known immune escape and tumor invasion mechanisms, revealing several druggable drivers of tumor progression in the patient under study. This proof-of-principle study highlights the potential of in-depth CN profiling in routine clinical samples to inform microenvironment-directed therapies. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Tancredi Massimo Pentimalli
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin
| | - Simon Schallenberg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Daniel León-Periñán
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ivano Legnini
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Human Technopole, Milan, Italy
| | - Ilan Theurillat
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Gwendolin Thomas
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anastasiya Boltengagen
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sonja Fritzsche
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany; Humboldt-Universität zu Berlin, Institute of Biology, 10099 Berlin, Germany
| | - Jose Nimo
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany; Humboldt-Universität zu Berlin, Institute of Biology, 10099 Berlin, Germany
| | | | - Gabriel Dernbach
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany; Aignostics GmbH, Berlin, Germany; BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| | | | | | | | - Yan Liang
- NanoString® Technologies, Inc, Seattle, WA, USA
| | | | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Padua, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Fabian Coscia
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Andrew Woehler
- Systems Biology Imaging Platform, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA
| | - Nikos Karaiskos
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Frederick Klauschen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany; Institute of Pathology, Ludwig Maximilians Universität, Munich, Germany
| | - Nikolaus Rajewsky
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany; NeuroCure Cluster of Excellence, Berlin, Germany; German Cancer Consortium (DKTK), Berlin, Germany; National Center for Tumor Diseases (NCT), Site Berlin, Berlin, Germany.
| |
Collapse
|
26
|
Fu L, Xie F, Sun P, Dong Y, Zhou K, Jiang L, Wu R, Han Y, Wu H, Tang G, Zhou W. First clinical investigation to predict lymphovascular and/or perineural invasion in gastric cancer using 18F-FAPI-42 PET/CT parameters. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07325-9. [PMID: 40387910 DOI: 10.1007/s00259-025-07325-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/28/2025] [Indexed: 05/20/2025]
Abstract
OBJECTIVE This study was conducted to explore the predictive value of PET parameters derived from 18F-FAPI-42 PET/CT in assessing lymphovascular and/or perineural invasion (LVI/PNI) in gastric cancer (GC) patients. METHODS 72 GC patients who underwent 18F-FAPI-42 PET/CT prior to surgical resection were included. Clinicopathological factors and PET parameters were collected and analyzed in LVI/PNI-negative and LVI/PNI-positive groups. The predictive value of PET parameters for LVI/PNI status was evaluated using the receiver operating characteristic (ROC) curve. A nomogram was developed using significant predictors from multivariate stepwise regression analysis and its performance was assessed by decision curve analysis (DCA). RESULTS Univariate analysis indicated a significant association between LVI/PNI status and PET parameters (SUVmax, SUVmean, and TBR) (all p < 0.001). The area under the ROC curve (AUC) values for predicting LVI/PNI were 0.932 [95% CI (0.877-0.987)] for SUVmax, 0.923 [95% CI (0.861-0.984)] for SUVmean, and 0.925 [95% CI (0.865-0.985)] for TBR. The optimal cutoff values for prediction, along with their corresponding sensitivity and specificity, were 3.86 (93.3% and 81.5%) for SUVmax, 2.04 (93.3% and 81.5%) for SUVmean, and 9.75 (91.1% and 81.5%) for TBR. Multivariate analysis identified histological grade and SUVmax as independent risk factors for LVI/PNI prediction. Our nomogram had good discriminatory ability (AUC = 0.934) and offered net benefits in predicting LVI/PNI status by DCA. CONCLUSION This study demonstrates that FAPI uptake parameters exhibit an exceptionally high capacity and serve as a noninvasive preoperative tool for predicting LVI/PNI status in GC, with SUVmax emerging as the most suitable predictive indicator.
Collapse
Affiliation(s)
- Lilan Fu
- Department of Nuclear Medicine, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Nuclear Medicine, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Fei Xie
- Department of Nuclear Medicine, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Penghui Sun
- Department of Nuclear Medicine, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ye Dong
- Department of Nuclear Medicine, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kemin Zhou
- Department of Nuclear Medicine, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Li Jiang
- Department of Nuclear Medicine, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ruihe Wu
- Department of Nuclear Medicine, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yanjiang Han
- Department of Nuclear Medicine, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hubing Wu
- Department of Nuclear Medicine, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Ganghua Tang
- Department of Nuclear Medicine, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Wenlan Zhou
- Department of Nuclear Medicine, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
27
|
Jaa A, Buque A. Novel metabolic routes to cancer immune evasion. Trends Cancer 2025:S2405-8033(25)00125-6. [PMID: 40393916 DOI: 10.1016/j.trecan.2025.05.002] [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: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025]
Abstract
The tumor microenvironment (TME) comprises heterogeneous cell types that closely interact with each other. Crosstalk among the TME components determines antitumor immune responses and their sensitivity to therapies such as immunotherapy. Recent studies published in Cancer Cell by Tang et al. and Zhu et al. identify two novel metabolic adaptations that tumors use to facilitate immune evasion. These targetable mechanisms suggest new avenues to improve antitumor immune responses.
Collapse
Affiliation(s)
- Ayoub Jaa
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Aitziber Buque
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Zhao Z, Li Q, Qu C, Jiang Z, Jia G, Lan G, Luan Y. A collagenase nanogel backpack improves CAR-T cell therapy outcomes in pancreatic cancer. NATURE NANOTECHNOLOGY 2025:10.1038/s41565-025-01924-1. [PMID: 40389641 DOI: 10.1038/s41565-025-01924-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/31/2025] [Indexed: 05/21/2025]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized the treatment of haematological malignancies. Challenges in overcoming physical barriers however greatly limit CAR-T cell efficacy in solid tumours. Here we show that an approach based on collagenase nanogel generally improves the outcome of T cell-based therapies, and specifically of CAR-T cell therapy. The nanogels are created by cross-linking collagenase and subsequently modifying them with a CXCR4 antagonist peptide. These nanogels can bind CAR-T cells via receptor-ligand interaction, resulting in cellular backpack delivery systems. The nanogel backpacks modulate tumoural infiltration and localization of CAR-T cells by surmounting physical barriers and disrupting chemokine-mediated CAR-T cell imprisonment, thereby addressing their navigation deficiency within solid tumours. Our approach offers a promising strategy for pancreatic cancer therapy and holds potential for advancing CAR-T cell therapy towards clinical applications.
Collapse
Affiliation(s)
- Zhipeng Zhao
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qian Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chenghao Qu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Zeyu Jiang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guoqing Jia
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Gongde Lan
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuxia Luan
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Key Laboratory of Chemical Biology (Ministry of Education), Shandong Key Laboratory of Targeted Drug Delivery and Advanced Pharmaceutics, NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
29
|
Yang Y, Zhu L, Xu Y, Liang L, Liu L, Chen X, Li H, Liu H. The progress and prospects of targeting the adenosine pathway in cancer immunotherapy. Biomark Res 2025; 13:75. [PMID: 40390144 PMCID: PMC12090549 DOI: 10.1186/s40364-025-00784-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/26/2025] [Indexed: 05/21/2025] Open
Abstract
Despite the notable success of cancer immunotherapy, its effectiveness is often limited in a significant proportion of patients, highlighting the need to explore alternative tumor immune evasion mechanisms. Adenosine, a key metabolite accumulating in hypoxic tumor regions, has emerged as a promising target in oncology. Inhibiting the adenosinergic pathway not only inhibits tumor progression but also holds potential to enhance immunotherapy outcomes. Multiple therapeutic strategies targeting this pathway are being explored, ranging from preclinical studies to clinical trials. This review examines the complex interactions between adenosine, its receptors, and the tumor microenvironment, proposing strategies to target the adenosinergic axis to boost anti-tumor immunity. It also evaluates early clinical data on pharmacological inhibitors of the adenosinergic pathway and discusses future directions for improving clinical responses.
Collapse
Affiliation(s)
- Yuying Yang
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Lin Zhu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yantao Xu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Long Liang
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Li Liu
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Hui Li
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Hong Liu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
30
|
Shah S, D'Souza GGM. Modeling Tumor Microenvironment Complexity In Vitro: Spheroids as Physiologically Relevant Tumor Models and Strategies for Their Analysis. Cells 2025; 14:732. [PMID: 40422235 DOI: 10.3390/cells14100732] [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: 04/18/2025] [Revised: 05/11/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Drug delivery to solid tumors is challenged by multiple physiological barriers arising from the tumor microenvironment, including dense extracellular matrix, cellular heterogeneity, hypoxic gradients, and elevated interstitial fluid pressure. These features hinder the uniform distribution and accumulation of therapeutics, reducing treatment efficacy. Despite their widespread use, conventional two-dimensional monolayer cultures fail to reproduce these complexities, contributing to the poor translational predictability of many preclinical candidates. Three-dimensional multicellular tumor spheroids have emerged as more representative in vitro models that capture essential features of tumor architecture, stromal interactions, and microenvironmental resistance mechanisms. Spheroids exhibit spatially organized regions of proliferation, quiescence, and hypoxia, and can incorporate non-tumor cells to mimic tumor-stroma crosstalk. Advances in spheroid analysis now enable detailed evaluation of drug penetration, cellular migration, cytotoxic response, and molecular gradients using techniques such as optical and confocal imaging, large-particle flow cytometry, biochemical viability assays, and microfluidic integration. By combining physiological relevance with analytical accessibility, spheroid models support mechanistic studies of drug transport and efficacy under tumor-like conditions. Their adoption into routine preclinical workflows has the potential to improve translational accuracy while reducing reliance on animal models.
Collapse
Affiliation(s)
- Shrey Shah
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
- Atom Bioworks Inc., Cary, NC 27513, USA
| | - Gerard G M D'Souza
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
| |
Collapse
|
31
|
Wang H, Cheng P, Wang J, Lv H, Han J, Hou Z, Xu R, Chen W. Advances in spatial transcriptomics and its application in the musculoskeletal system. Bone Res 2025; 13:54. [PMID: 40379648 DOI: 10.1038/s41413-025-00429-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 05/19/2025] Open
Abstract
While bulk RNA sequencing and single-cell RNA sequencing have shed light on cellular heterogeneity and potential molecular mechanisms in the musculoskeletal system in both physiological and various pathological states, the spatial localization of cells and molecules and intercellular interactions within the tissue context require further elucidation. Spatial transcriptomics has revolutionized biological research by simultaneously capturing gene expression profiles and in situ spatial information of tissues, gradually finding applications in musculoskeletal research. This review provides a summary of recent advances in spatial transcriptomics and its application to the musculoskeletal system. The classification and characteristics of data acquisition techniques in spatial transcriptomics are briefly outlined, with an emphasis on widely-adopted representative technologies and the latest technological breakthroughs, accompanied by a concise workflow for incorporating spatial transcriptomics into musculoskeletal system research. The role of spatial transcriptomics in revealing physiological mechanisms of the musculoskeletal system, particularly during developmental processes, is thoroughly summarized. Furthermore, recent discoveries and achievements of this emerging omics tool in addressing inflammatory, traumatic, degenerative, and tumorous diseases of the musculoskeletal system are compiled. Finally, challenges and potential future directions for spatial transcriptomics, both as a field and in its applications in the musculoskeletal system, are discussed.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China
| | - Peng Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Juan Wang
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China
| | - Hongzhi Lv
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China
| | - Jie Han
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhiyong Hou
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China
| | - Ren Xu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cells, State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China.
| | - Wei Chen
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China.
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China.
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China.
| |
Collapse
|
32
|
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
|
33
|
Kim YS, Kim SJ. Diagnostic Performances of Radiolabeled FAPI PET/CT for Lymph Node Staging in Head and Neck Cancer Patients: Comparison With 18F-FDG PET/CT. Clin Nucl Med 2025:00003072-990000000-01726. [PMID: 40367495 DOI: 10.1097/rlu.0000000000005973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/14/2025] [Indexed: 05/16/2025]
Abstract
OBJECTIVE The purpose of the current study was to compare the diagnostic performances of radiolabeled FAPI and 18F-FDG PET/CT for the detection of lymph node (LN) metastasis in head and neck cancer (HNC) patients. METHODS The PubMed, Cochrane database, and EMBASE database, from the earliest available date of indexing through December 31, 2024, were searched for studies comparing diagnostic performances of radiolabeled FAPI and 18F-FDG PET/CT for the detection of metastatic LN in HNC patients. We estimated pooled sensitivities and specificities across studies. RESULTS Across 8 studies (14 results), the pooled sensitivity of FAPI PET/CT was 0.89 and the pooled specificity was 0.93. The pooled sensitivity of 18F-FDG PET/CT was 0.91 and the pooled specificity was 0.50. On patient-based analysis, the estimated sensitivity and specificity of FAPI were 0.96 and 0.96, and those of 18F-FDG were 0.95 and 0.34, respectively. On lesion-based analysis, the estimated sensitivity and specificity of FAPI were 0.84 and 0.94, and those of 18F-FDG were 0.86 and 0.78, respectively. On neck side-based analysis, the estimated sensitivity and specificity of FAPI were 0.88 and 0.79, and those of 18F-FDG were 0.91 and 0.29, respectively. CONCLUSIONS Radiolabeled FAPI showed a good diagnostic performance for the detection of metastatic LN in HNC patients. Also, 18F-FDG PET/CT revealed low specificity for LN staging in HNC patients. Future large multicenter research with more patients would be necessary to provide a more comprehensive overview of the usefulness of radiolabeled FAPI for LN staging in HNC patients.
Collapse
Affiliation(s)
- Yun Seong Kim
- Department of Internal Medicine
- BioMedical Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital
| | - Seong-Jang Kim
- BioMedical Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital
- Department of Nuclear Medicine, Pusan National University School of Medicine
- Department of Nuclear Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea
| |
Collapse
|
34
|
Zou AE, Kongthong S, Mueller AA, Brenner MB. Fibroblasts in immune responses, inflammatory diseases and therapeutic implications. Nat Rev Rheumatol 2025:10.1038/s41584-025-01259-0. [PMID: 40369134 DOI: 10.1038/s41584-025-01259-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2025] [Indexed: 05/16/2025]
Abstract
Once regarded as passive bystander cells of the tissue stroma, fibroblasts have emerged as active orchestrators of tissue homeostasis and disease. From regulating immunity and controlling tissue remodelling to governing cell growth and differentiation, fibroblasts assume myriad roles in guiding normal tissue development, maintenance and repair. By comparison, in chronic inflammatory diseases such as rheumatoid arthritis, fibroblasts recruit and sustain inflammatory leukocytes, become dominant producers of pro-inflammatory factors and catalyse tissue destruction. In other disease contexts, fibroblasts promote fibrosis and impair host control of cancer. Single-cell studies have uncovered striking transcriptional and functional heterogeneity exhibited by fibroblasts in both normal tissues and diseased tissues. In particular, advances in the understanding of fibroblast pathology in rheumatoid arthritis have shed light on pathogenic fibroblast states in other chronic diseases. The differentiation and activation of these fibroblast states is driven by diverse physical and chemical cues within the tissue microenvironment and by cell-intrinsic signalling and epigenetic mechanisms. These insights into fibroblast behaviour and regulation have illuminated therapeutic opportunities for the targeted deletion or modulation of pathogenic fibroblasts across many diseases.
Collapse
Affiliation(s)
- Angela E Zou
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Suppawat Kongthong
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alisa A Mueller
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA and Palo Alto Veterans Affairs Health Care System, Palo Alto, CA, USA
| | - Michael B Brenner
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
35
|
Faa G, Ziranu P, Pretta A, Cau F, Castagnola M, Spanu D, Saba G, D'Agata AP, Tiwari E, Suri JS, Scartozzi M, Saba L. Cancer-associated fibroblasts (CAFs) and plaque-associated fibroblasts (PAFs): Unraveling the cellular crossroads of atherosclerosis and cancer. Biomed Pharmacother 2025; 188:118145. [PMID: 40373629 DOI: 10.1016/j.biopha.2025.118145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/17/2025] Open
Abstract
Atherosclerosis is a complex process involving various cells and molecules within the atherosclerotic plaque. Recent evidence suggests that plaque-associated fibroblasts (PAFs), also known as atherosclerosis-associated fibroblasts (AAFs), might play a significant role in the development and progression of the disease. The microenvironment of the atherosclerotic plaque, resembling the tumor microenvironment (TME), includes various cellular populations like plaque-associated macrophages (PAMs), plaque-associated neutrophils (PANs), vascular smooth muscle cells (VSMCs), myeloid-derived suppressor cells (MDSCs), and PAFs. Similar to cancer-associated fibroblasts (CAFs) in tumors, PAFs exhibits a wide range of characteristics and functions. Their interactions with endothelial cells, smooth muscle cells, and other stromal cells, including adventitial fibroblast precursors, significantly influence atherosclerosis progression. Moreover, the ability of PAFs to express various markers such as alpha-SMA, Desmin, VEGF, and GFAP, highlights their diverse origins from different precursor cells, including vascular smooth muscle cells, endothelial cells, glial cells of the enteric nervous system, adventitial fibroblast precursors, as well as resident and circulating fibrocytes. This article explores the molecular interactions between PAFs, cells associated with atherosclerosis, and other stromal cells. It further examines the role of PAFs in the development and progression of atherosclerosis, and compares their features with those of CAFs. The research suggests that studying tumor-associated fibroblasts can help understand fibroblast subpopulations in atherosclerotic plaque. Identifying specific subpopulations could provide new insight into atherosclerosis complexity and lead to the development of innovative drugs for medical intervention.
Collapse
Affiliation(s)
- Gavino Faa
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy.
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Flaviana Cau
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Massimo Castagnola
- Laboratory of Proteomics, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Dario Spanu
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Giorgio Saba
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Alessandra Pia D'Agata
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Ekta Tiwari
- Department of Innovation. Global Biomedical Technologies, Inc., Roseville, CA 95661, USA
| | - Jasjit S Suri
- Department of ECE, Idaho State University, Pocatello, ID, 83209, USA; Department of CE, Graphics Era Deemed to be University, Dehradun 248002, India; University Center for Research & Development, Chandigarh University, Mohali, India; Symbiosis Institute of Technology, Nagpur Campus, Symbiosis International (Deemed University), Pune, INDIA; Stroke Diagnostic and Monitoring Division, AtheroPoint, Roseville, CA 95661, USA
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Luca Saba
- Department of Medical Sciences and Public Health, Unit of Radiology, University fo Cagliari, Cagliari, Italy
| |
Collapse
|
36
|
Taief KA, Nemec S, Middleton IA, Kilian KA, Thordarson P. Scrambled RGD Hexameric Peptide Hydrogel Supports Efficient Self-Assembly and Cell Activity. Chemistry 2025; 31:e202404410. [PMID: 40192287 PMCID: PMC12080297 DOI: 10.1002/chem.202404410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/22/2025]
Abstract
The amino acid sequence is crucial in controlling peptide-based hydrogel formation, whereby changing the position of a single amino acid can significantly alter the gel's properties. Herein, we report the gelation kinetics and cell viability of scrFmoc-GFFRDG (where we have scrambled the RGD-based gel hexapeptide; Fmoc-GFFRGD). The scrambled sequence showed improved gelation properties compared to the original Fmoc-GFFRGD sequence, with scrFmoc-GFFRDG forming a gel in under 10 min, significantly faster than the 2-h gelation time, and at a concentration eight times lower than the original Fmoc-GFFRGD sequence. We also examined the combination of the two gelators in a ratio of 1:1, final concentration of 0.4% (w/v). Interestingly, the stiffness of the hybrid hydrogel was ∼3 kPa, whereas individually, neither gelator at the same concentration exceeded 0.5 kPa. The cell-adhesion motif RGD improves the ability of the peptides to promote attachment of cells due to integrin recognition. However, when fibroblasts were cultured on the hydrogels, scrFmoc-GFFRDG yielded a higher level of α-SMA expression in cells than those cultured on Fmoc-GFFRGD, suggesting a microenvironment conducive to myofibroblast transitions. This study provides a new outlook on how a well-known scrambled peptide motif (RDG) can fine-tune hydrogel assembly and cell culture applications.
Collapse
Affiliation(s)
- Karrar Al Taief
- School of ChemistryUniversity of New South WalesSydneyNSW2052Australia
- The UNSW RNA InstituteUniversity of New South WalesSydneyNSW2052Australia
| | - Stephanie Nemec
- School of Materials Science and EngineeringUniversity of New South WalesSydneyNSW2052Australia
- Australian Centre for NanoMedicineUniversity of New South WalesSydneyNSW2052Australia
| | - Isis A. Middleton
- School of ChemistryUniversity of New South WalesSydneyNSW2052Australia
- The UNSW RNA InstituteUniversity of New South WalesSydneyNSW2052Australia
| | - Kristopher A. Kilian
- School of ChemistryUniversity of New South WalesSydneyNSW2052Australia
- School of Materials Science and EngineeringUniversity of New South WalesSydneyNSW2052Australia
- Australian Centre for NanoMedicineUniversity of New South WalesSydneyNSW2052Australia
| | - Pall Thordarson
- School of ChemistryUniversity of New South WalesSydneyNSW2052Australia
- The UNSW RNA InstituteUniversity of New South WalesSydneyNSW2052Australia
- Australian Centre for NanoMedicineUniversity of New South WalesSydneyNSW2052Australia
| |
Collapse
|
37
|
Choi J, Kwak Y, Park M, Jo JY, Kang JH, Myeong-Cherl K, Kim HR, Kim G, Kong SH, Park DJ, Lee HS, Lee HJ, Kim JM, Kim SG, Yang HK, Ryu JK, Cho SJ. Cancer-associated fibroblast-derived fibulin-5 promotes epithelial-mesenchymal transition in diffuse-type gastric cancer via cAMP response element-binding protein pathway, showing poor prognosis. Exp Mol Med 2025:10.1038/s12276-025-01447-8. [PMID: 40369121 DOI: 10.1038/s12276-025-01447-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/27/2025] [Accepted: 02/23/2025] [Indexed: 05/16/2025] Open
Abstract
Diffuse-type gastric cancer (DGC), characterized by poorly cohesive cells within fibrotic stroma, is associated with advanced disease and poor prognosis. Here, to identify distinct biomarkers for DGC compared with intestinal-type gastric cancer, we constructed a comprehensive large-scale signaling network using RNA-sequencing data from three genomic databases (The Cancer Genome Atlas, GSE62254 and GSE26253), developed a mathematical model and conducted simulation analyses. For validation, we used tissue microarray blocks of gastric cancers with immunohistochemical staining, single-cell RNA sequencing, primary cultures of cancer-associated fibroblasts (CAFs) and organoids, and a co-culture system involving CAFs and cancer cells. Signaling network analysis identified six differentially activated signaling components across the database, including BIRC5, TTK, NEK2, FHL1, NR2F1 and FBLN5. Among the differentially activated signaling components, high tumoral expression of fibulin-5 protein encoded by FBLN5 correlated with poor overall and disease-specific survival rates in patients with DGC, even after adjusting for the tumor, node, metastases (TNM) stage. Fibulin-5, derived from CAFs within DGC stroma, promoted organoid growth and epithelial-mesenchymal transition (EMT) in DGC cell lines via the cAMP response element-binding protein (CREB) pathway in a CAF co-culture system. FBLN5 knockdown in CAFs reduced the aggressive phenotype of co-cultured DGC cells, while CREB inhibitors reversed EMT. Furthermore, levels of secreted FBLN5 in patient blood samples correlated with its expression in primary tumors. In summary, fibulin-5 secreted by CAFs and interacted with DGC cells promotes EMT and is clinically associated with poor patient outcomes. These findings suggest fibulin-5 as a potential prognostic marker and therapeutic target in patients with DGC.
Collapse
Affiliation(s)
- Jinju Choi
- Division of Gastroenterology, Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Yoonjin Kwak
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Miree Park
- Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jeong Yeon Jo
- Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jun Hyuk Kang
- Center for Gastric Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Kook Myeong-Cherl
- Center for Gastric Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Hang-Rae Kim
- Department of Biomedical Sciences, BK21 FOUR Biomedical Science Project, and Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Gwanghun Kim
- Department of Biomedical Sciences, BK21 FOUR Biomedical Science Project, and Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seong-Ho Kong
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Do-Joong Park
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyuk-Joon Lee
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jung Mogg Kim
- Department of Microbiology, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Sang Gyun Kim
- Division of Gastroenterology, Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Han-Kwang Yang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji Kon Ryu
- Division of Gastroenterology, Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Soo-Jeong Cho
- Division of Gastroenterology, Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
38
|
Luo Y, Fu H, Yu C. Based on small molecules: development and application of fibroblast activation protein inhibitors radiopharmaceutical in tumor precision therapy. Front Pharmacol 2025; 16:1593380. [PMID: 40438601 PMCID: PMC12116444 DOI: 10.3389/fphar.2025.1593380] [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: 03/14/2025] [Accepted: 04/30/2025] [Indexed: 06/01/2025] Open
Abstract
The discovery of biomarkers for malignant tumors is driving the development of new radiopharmaceuticals in nuclear medicine. The development and optimization of novel radiopharmaceuticals to occupy an increasingly important role in tumor diagnosis and treatment. In recent years, fibroblast activation protein (FAP) has gained attention as a promising tumor target due to its widespread expression across various tumors. FAP inhibitor (FAPI) radiopharmaceuticals are considered to be the most promising to be developed for targeting FAP due to their rapid and specific tumor targeting. This review briefly outlines the developmental history of FAP-targeted small-molecule enzyme activity inhibitors, highlighting the effective role of targeting molecules, linkers, and certain functional groups in the delivery of radioisotopes to cancerous tissues. These development strategies will serve as a reference for the further development and application of relevant radiopharmaceuticals. This review also delineates the progress on clinical FAPI as a radioisotope delivery vehicle for the targeted radioligand therapy of tumors and introduces the latest combination therapy involving FAPI radiopharmaceutical for tumor treatment. The findings provide novel therapeutic insights into the targeted radioligand therapy of tumors.
Collapse
Affiliation(s)
- Yihui Luo
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Haitian Fu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Chunjing Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
39
|
Bains RS, Raju TG, Semaan LC, Block A, Yamaguchi Y, Priceman SJ, George SC, Shirure VS. Vascularized tumor-on-a-chip to investigate immunosuppression of CAR-T cells. LAB ON A CHIP 2025; 25:2390-2400. [PMID: 40289711 DOI: 10.1039/d4lc01089b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Chimeric antigen receptor (CAR)-T cell immunotherapy, effective in blood cancers, shows limited success in solid tumors, such as prostate, pancreatic, and brain cancers due, in part, to an immunosuppressive tumor microenvironment (TME). Immunosuppression affects various cell types, including tumor cells, macrophages, and endothelial cells. Conventional murine-based models offer limited concordance with human immunology and cancer biology. Therefore, we have developed a human "tumor-on-a-chip" (TOC) platform to model elements of immunosuppression at high spatiotemporal resolution. Our TOC features an endothelial cell-lined channel that mimics features of an in vivo capillary, such as cell attachment and extravasation across the endothelium and into the TME. Using 70 kDa dextran and fluorescence-recovery-after-photobleaching (FRAP), we confirmed physiologic interstitial flow velocities (0.1-1 μm s-1). Our device demonstrates that tumor-derived factors can diffuse in the opposite direction of interstitial flow to reach the endothelium up to 200 μm away, and at concentrations as high as 20% of those at the tumor margin. M2-like immunosuppressive macrophages and endothelial cells affect prostate tumor cell growth, clustering, and migration. M2-like macrophages also induce PD-L1 and inhibit ICAM-1 gene expression on the adjacent endothelium in a pattern that limits CAR-T cell extravasation and effector function. This observation is abrogated in the presence of the anti-PD-L1 drug atezolizumab. These results provide mechanistic insight for in vivo observations showing limited CAR-T cell extravasation and effector function in solid tumors. Furthermore, they point to a specific role of M2 macrophages in driving CAR-T cell migration into and within the TME and could prove useful in the development of novel therapies to improve solid tumor CAR-T cell therapies.
Collapse
Affiliation(s)
- Rajul S Bains
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, 95616, USA.
| | - Tara G Raju
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, 95616, USA.
| | - Layla C Semaan
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, 95616, USA.
| | - Anton Block
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, 95616, USA.
| | - Yukiko Yamaguchi
- Department of Medicine, KSOM/NCCC Center for Cancer Cellular Immunotherapy Research, University of Southern California, Los Angeles, CA, 90098, USA
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, USA
| | - Saul J Priceman
- Department of Medicine, KSOM/NCCC Center for Cancer Cellular Immunotherapy Research, University of Southern California, Los Angeles, CA, 90098, USA
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, 91010, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, 95616, USA.
- Department of Surgery, University of California, Davis,, Sacramento, CA, 95817, USA
| | - Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
40
|
Kaleem M, Azmi L, Shahzad N, Taha M, Kumar S, Mujtaba MA, Hazazi AAH, Kayali A. Epigenetic dynamics and molecular mechanisms in oncogenesis, tumor progression, and therapy resistance. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04217-5. [PMID: 40358685 DOI: 10.1007/s00210-025-04217-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025]
Abstract
Cancer progression is governed by a dynamic interplay of genetic, epigenetic, and molecular mechanisms that regulate tumor initiation, growth, metastasis, and therapy resistance. This review highlights key molecular pathways involved in oncogenesis, focusing on genetic alterations (mutations, amplifications, and translocations) in oncogenes (RAS and MYC) and tumor suppressor genes (TP53 and PTEN). Additionally, genomic instability, resulting from defective DNA repair mechanisms like mismatch repair and homologous recombination (HR), is identified as a critical factor contributing to tumor heterogeneity and clonal evolution. Epigenetic modifications, including DNA methylation, histone acetylation, and non-coding RNA regulation, further remodel chromatin structure and modulate gene expression, influencing tumor initiation, growth, metastasis, and response to treatment. Post-translational modifications, such as the attachment of a Small Ubiquitin-like Modifier (SUMO) to a target protein and ubiquitination, further influence autophagy, apoptosis, and cellular plasticity, enabling cancer cells to survive therapeutic stress. Cutting-edge technologies such as CRISPR-Cas9-mediated epigenome editing and single-cell RNA sequencing have opened new doors to understanding cellular diversity and regulatory networks in cancer. The review further examines the tumor microenvironment, including stromal remodeling, immune evasion, and hypoxia-driven signaling pathways, which are critical modulators of tumor progression and drug resistance to treatment. By integrating molecular, genetic, and epigenetic perspectives, this study underscores the crucial need for innovative, targeted therapeutic approaches to address the complexity and adaptability of cancer, thereby paving the way for more effective treatments.
Collapse
Affiliation(s)
- Mohammed Kaleem
- Department of Pharmacology, Dadasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, Maharashtra, India
| | - Lubna Azmi
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Science, University of Lucknow, Uttar Pradesh, Lucknow, India
| | - Naiyer Shahzad
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Murtada Taha
- Department of Clinical Laboratory Science, Prince Sultan Military College of Health Sciences, Dhahran, Saudi Arabia
| | - Shiv Kumar
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh, Varanasi, India
| | - Md Ali Mujtaba
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Arar, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| | | | - Asaad Kayali
- Department of Health Sciences, Higher Colleges of Technology, Al Ain, United Arab Emirates
| |
Collapse
|
41
|
Zhao J, Liu M, Zhu C, Li Z, Liu Z, Abulizi D, Liu S, Wang X, Yang H, Hou X. Cancer-associated fibroblasts and metabolic reprogramming predict pathologic response to neoadjuvant PD-1 blockade in resected non-small cell lung cancer. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01067-4. [PMID: 40358847 DOI: 10.1007/s13402-025-01067-4] [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: 02/14/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
PURPOSE Immunotherapy has transformed the neoadjuvant treatment landscape for patients with resectable locally advanced non-small cell lung cancer (NSCLC). However, a population of patients cannot obtain major pathologic response (MPR) and thus benefit less from neoadjuvant immunotherapy, highlighting the need to uncover the underlying mechanisms driving resistance to immunotherapy. METHODS Two published single-cell RNA sequencing (scRNA-seq) datasets were used to analyze the subsets of cancer-associated fibroblasts (CAFs) and T cells and functional alterations after neoadjuvant immunotherapy. The stromal signature predicting ICI response was identified and validated using our local cohort with stage III NSCLC receiving neoadjuvant immunotherapy and other 4 public ICI transcriptomic cohorts. RESULTS Non-MPR tumors showed higher enrichment of CAFs and increased extracellular matrix deposition than MPR tumors, as suggested by bioinformatic analysis. Further, CAF-mediated immune suppression may involve reciprocal interactions with T cells in addition to a physical barrier mechanism. In contrast, MPR tumors demonstrated therapy-induced activation of memory CD8+ T cells into an effector phenotype. Additionally, neoadjuvant immunotherapy resulted in expansion of precursor exhausted T (Texp) cells, which were remodeled into an anti-tumor phenotype. Notably, we identified metabolic heterogeneity within distinct T cell clusters during immunotherapy. Methionine recycling emerged as a predictive factor for T-cell differentiation and a favorable pathological response. The stromal signature was associated with ICI response, and this association was validated in five independent ICI transcriptomic cohorts. CONCLUSION These discoveries underscore the distinct tumor microenvironments in MPR and non-MPR patients and may elucidate resistance mechanisms to immunotherapy in NSCLC.
Collapse
Affiliation(s)
- Jiaqi Zhao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou City, Guangdong Province, 510060, PR China
| | - Maolin Liu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou City, Guangdong Province, 510060, PR China
| | - Chongmei Zhu
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Zhuolin Li
- Guangzhou BioScript Biotechnology Co., Ltd, Guangzhou, PR China
| | - Zuhui Liu
- The Department of Breast Disease, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, PR China
| | - Dilimulati Abulizi
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou City, Guangdong Province, 510060, PR China
| | - Siqing Liu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou City, Guangdong Province, 510060, PR China
| | - Xin Wang
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou City, Guangdong Province, 510060, PR China
| | - Haoxian Yang
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou City, Guangdong Province, 510060, PR China.
| | - Xue Hou
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou City, Guangdong Province, 510060, PR China.
| |
Collapse
|
42
|
Wang M, Pu N, Bo X, Chen F, Zhou Y, Cheng Q. Significance and mechanisms of perineural invasion in malignant tumors. Front Oncol 2025; 15:1572396. [PMID: 40421086 PMCID: PMC12104087 DOI: 10.3389/fonc.2025.1572396] [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: 02/07/2025] [Accepted: 04/18/2025] [Indexed: 05/28/2025] Open
Abstract
Cancer remains the second leading cause of death worldwide. Tumor invasion and metastasis pose significant challenges for clinical management. In addition to the traditional pathways of metastasis such as hematologic or lymphatic transmission, perineural invasion (PNI) has become a unique mechanism of metastasis, which is closely associated with neuropathic pain, motor deficits, and poor prognosis. PNI is often observed in malignant tumors of the pancreas, head and neck, gastrointestinal tract, and lungs, and it reflects a unique neurotropic transfer behavior utilizing neural networks. Despite its clinical significance, targeted therapies for PNI are still lacking. This review synthesizes current evidences regarding PNI, elucidates the clinical significance of PNI in tumor metastasis, prognosis, and neurological dysfunction. By integrating the latest advances in multi-omics, we analyzed the potential key molecular pathways and tumor microenvironment drivers of PNI, and proposed future research directions for developing PNI-specific therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Mengyao Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Niu Pu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Xitong Bo
- Department of Surgery, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Fuxiang Chen
- Department of Surgery, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Yilong Zhou
- Department of Surgery, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Qiong Cheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| |
Collapse
|
43
|
Zhu H, Wang J, Liu Y, Wang X, Lu TJ, Xu F, Lin M. Estrogen attenuates stiffness-driven fibrotic signaling via transcriptional regulation. Biophys J 2025:S0006-3495(25)00286-3. [PMID: 40364520 DOI: 10.1016/j.bpj.2025.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 04/19/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025] Open
Abstract
Fibrosis, marked by excessive extracellular matrix (ECM) accumulation, underlies functional decline in numerous diseases and often presents with sex-specific differences in severity. Although biochemical pathways have been widely studied, the contribution of mechanical cues-particularly ECM stiffness-to these disparities remains unclear. Here, we develop an integrative mechanobiological model to investigate how estrogen modulates stiffness-mediated fibrotic progression. The model reveals that ECM stiffness activates fibroblasts through two key pathways: a rapid nuclear translocation of mechanosensitive factors (MRTF and TAZ) and a delayed transforming growth factor β/Smad cascade, both of which enhance α-smooth muscle actin expression and matrix production. Moreover, we uncover a stiffness-induced "mechanical memory" effect, maintained through a miR-21/Smad feedback loop that sustains fibrotic signaling even after stiffness reduction. Estrogen, acting via estrogen receptor α, counteracts this process by promoting Smad degradation and interrupting the feedback loop, thereby dampening fibrosis. This work offers new insight into the mechanochemical regulation of sex-biased fibrosis and points to potential sex-specific therapeutic targets.
Collapse
Affiliation(s)
- Hongyuan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jin Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Out-patient Department, School of Stomatology, The Fourth Military Medical University, Xi'an, P.R. China
| | - Xiaohong Wang
- Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, Xi'an, P.R. China
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, P.R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China.
| |
Collapse
|
44
|
Zhang YP, Guo ZQ, Cai XT, Rong ZX, Fang Y, Chen JQ, Zhuang KM, Ruan MJ, Ma SC, Lin LY, Han DD, Li YS, Wang YY, Wang J, Cao CH, Tang XR, Xie QK, Chen Y, Lin Y, Tan JL, Yu ZH, Wu ZN, Wei W, Zheng DY, Zeng YJ, Ruan YC, Xu ZP, Gu JZ, Xiao LS, Liu L, Guan J, Bai X, Wu DH, Dong ZY. PAI-1-driven SFRP2 high cancer-associated fibroblasts hijack the abscopal effect of radioimmunotherapy. Cancer Cell 2025; 43:856-874.e9. [PMID: 40086438 DOI: 10.1016/j.ccell.2025.02.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/15/2024] [Accepted: 02/20/2025] [Indexed: 03/16/2025]
Abstract
The abscopal effect of radioimmunotherapy, wherein tumor shrinkage occurs beyond the irradiated field, is therapeutically promising but clinically rare. The mechanisms underlying this effect remain elusive. Here, in vivo genome-wide CRISPR screening identifies SFRP2 as a potential stromal regulator of the abscopal effect. SFRP2 exhibits cancer-associated fibroblast (CAF)-specific expression and radioimmunotherapy-mediated upregulation in unirradiated tumors. Conditional Sfrp2 knockout in CAFs boosts the abscopal effect by rewiring the vascular-immune microenvironment to promote CD8+ T cell recruitment to unirradiated tumors. In vivo lineage tracing reveals that elevated SFRP2 correlates with radioimmunotherapy-driven pericyte lineage commitment. Serum proteomics reveals that irradiated-tumor-secreted PAI-1 triggers distant tumor pericyte cell-fate transition into SFRP2high CAFs via the LRP1/p65 axis. Pharmacologically blocking SFRP2 or PAI-1 enhances the abscopal effect in humanized patient-derived xenograft models. Our findings collectively illustrate that PAI-1-induced SFRP2high CAFs serve as critical stromal regulator to hijack the abscopal effect, providing promising targets for enhancing radioimmunotherapy effectiveness.
Collapse
Affiliation(s)
- Yan-Pei Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ze-Qin Guo
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Ting Cai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zi-Xuan Rong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuan Fang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jia-Qi Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kui-Mao Zhuang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Min-Jie Ruan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Si-Cong Ma
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Le-Yi Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Duan-Duan Han
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yang-Si Li
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yuan-Yuan Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jian Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chuan-Hui Cao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xin-Ran Tang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qian-Kun Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yue Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yan Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jia-Le Tan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zi-Hang Yu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ze-Nan Wu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wei Wei
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, Hubei 441000, China
| | - Da-Yong Zheng
- Department of Oncology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Shunde 528333, China
| | - Yu-Jie Zeng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Ying-Chen Ruan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zi-Peng Xu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jun-Zi Gu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lu-Shan Xiao
- Big Data Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Li Liu
- Big Data Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Health Management Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jian Guan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Xue Bai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - De-Hua Wu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Zhong-Yi Dong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
45
|
Liu Y, Sinjab A, Min J, Han G, Paradiso F, Zhang Y, Wang R, Pei G, Dai Y, Liu Y, Cho KS, Dai E, Basi A, Burks JK, Rajapakshe KI, Chu Y, Jiang J, Zhang D, Yan X, Guerrero PA, Serrano A, Li M, Hwang TH, Futreal A, Ajani JA, Solis Soto LM, Jazaeri AA, Kadara H, Maitra A, Wang L. Conserved spatial subtypes and cellular neighborhoods of cancer-associated fibroblasts revealed by single-cell spatial multi-omics. Cancer Cell 2025; 43:905-924.e6. [PMID: 40154487 PMCID: PMC12074878 DOI: 10.1016/j.ccell.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/09/2024] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are a multifaceted cell population essential for shaping the tumor microenvironment (TME) and influencing therapy responses. Characterizing the spatial organization and interactions of CAFs within complex tissue environments provides critical insights into tumor biology and immunobiology. In this study, through integrative analyses of over 14 million cells from 10 cancer types across 7 spatial transcriptomics and proteomics platforms, we discover, validate, and characterize four distinct spatial CAF subtypes. These subtypes are conserved across cancer types and independent of spatial omics platforms. Notably, they exhibit distinct spatial organizational patterns, neighboring cell compositions, interaction networks, and transcriptomic profiles. Their abundance and composition vary across tissues, shaping TME characteristics, such as levels, distribution, and state composition of tumor-infiltrating immune cells, tumor immune phenotypes, and patient survival. This study enriches our understanding of CAF spatial heterogeneity in cancer and paves the way for novel approaches to target and modulate CAFs.
Collapse
Affiliation(s)
- Yunhe Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ansam Sinjab
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jimin Min
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guangchun Han
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Francesca Paradiso
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuanyuan Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ruiping Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guangsheng Pei
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yibo Dai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX 77030, USA
| | - Yang Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kyung Serk Cho
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Enyu Dai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Akshay Basi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jared K Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kimal I Rajapakshe
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yanshuo Chu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jiahui Jiang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Daiwei Zhang
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xinmiao Yan
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Paola A Guerrero
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alejandra Serrano
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tae Hyun Hwang
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Luisa M Solis Soto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amir A Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX 77030, USA.
| | - Anirban Maitra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX 77030, USA; The James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
46
|
Li M, Cui Y, Yan Y, Zhao J, Lin X, Liu Q, Dong S, Nie M, Huang Y, Li B, Yin Y. Dual energy CT-derived quantitative parameters and hematological characteristics predict pathological complete response in neoadjuvant chemoradiotherapy esophageal squamous cell carcinoma patients. BMC Gastroenterol 2025; 25:357. [PMID: 40349002 PMCID: PMC12065240 DOI: 10.1186/s12876-025-03964-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 04/30/2025] [Indexed: 05/14/2025] Open
Abstract
PURPOSE There is no gold standard method to predict pathological complete response (pCR) in esophageal squamous cell carcinoma (ESCC) patients before surgery after neoadjuvant chemoradiotherapy (nCRT). This study aims to investigate whether dual layer detector dual energy CT (DECT) quantitative parameters and clinical features could predict pCR for ESCC patients after nCRT. PATIENTS AND METHODS This study retrospective recruited local advanced ESCC patients who underwent nCRT followed by surgical treatment from December 2019 to May 2023. According to pCR status (no visible cancer cells in primary cancer lesion and lymph nodes), patients were categorized into pCR group (N = 25) and non-pCR group (N = 28). DECT quantitative parameters were derived from conventional CT images, different monoenergetic (MonoE) images, virtual non-contrast (VNC) images, Z-effective (Zeff) images, iodine concentration (IC) images and electron density (ED) images. Slope of spectral curve (λHU), normalized iodine concentration (NIC), arterial enhancement fraction (AEF) and extracellular volume (ECV) were calculated. Difference tests and spearman correlation were used to select quantitative parameters for DECT model building. Multivariate logistic analysis was used to build clinical model, DECT model and combined model. RESULTS A total of 53 patients with locally advanced ESCC were enrolled in this study who received nCRT combined with surgery and underwent DECT examination before treatment. After spearman correlation analysis and multivariate logistic analysis, AEF and ECV showed significant roles between pCR and non-pCR groups. These two quantitative parameters were selected for DECT model. Multivariate logistic analysis revealed that LMR and RBC were also independent predictors in clinical model. The combined model showed the highest sensitivity, specificity, PPV and NPV compared to the clinical and DECT model. The AUC of the combined model is 0.893 (95%CI: 0.802-0.983). Delong's test revealed the combined model significantly different from clinical model (Z =-2.741, P = 0.006). CONCLUSION Dual-layer DECT derived ECV fraction and AEF are valuable predictors for pCR in ESCC patients after nCRT. The model combined DECT quantitative parameters and clinical features might be used as a non-invasive tool for individualized treatment decision of those ESCC patients. This study validates the role of DECT in pCR assessment for ESCC and a large external cohort is warranted to ensure the robustness of the proposed DECT evaluation criteria.
Collapse
Affiliation(s)
- Miaomiao Li
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China
- Shandong Medical College, Jinan, Shandong, China
| | - Yongbin Cui
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yuanyuan Yan
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Junfeng Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xinjun Lin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qianyu Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shushan Dong
- Clinical Science, Philips Healthcare, Beijing, China
| | - Mingming Nie
- Clinical Science, Philips Healthcare, Beijing, China
| | - Yong Huang
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Baosheng Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Yong Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
47
|
Mitra P, Saha U, Stephen KJ, Prasad P, Jena S, Patel AK, Bv H, Mondal SK, Kurkalang S, Roy S, Ghosh A, Roy SS, Das Sarma J, Biswas NK, Acharya M, Sharan R, Arun P, Jolly MK, Maitra A, Singh S. Tie2 activity in cancer associated myofibroblasts serves as novel target against reprogramming of cancer cells to embryonic-like cell state and associated poor prognosis in oral carcinoma patients. J Exp Clin Cancer Res 2025; 44:142. [PMID: 40349056 PMCID: PMC12065280 DOI: 10.1186/s13046-025-03405-8] [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: 01/05/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Myofibroblastic cancer-associated fibroblasts (CAF) in tumor stroma serves as an independent poor prognostic indicator, supporting higher stemness in oral cancer; however, the underlying biology is not fully comprehended. Here, we have explored the crucial role of Tunica Interna Endothelial Cell Kinase (Tie2/TEK) signaling in transition and maintenance of myofibroblastic phenotype of CAFs, and as possible link with the poor prognosis of head and neck squamous cell carcinoma (HNSCC) patients. METHODS Bulk and single cell RNA-sequencing (scRNAseq) methods and in-depth bioinformatic analysis were applied for CAF and cancer cells co-culture for studying molecular relationships. In vitro 3D-spheroid-forming ability, expression of stemness markers, in vivo tumor formation ability in zebrafish embryo and syngeneic mouse allografts formation was conducted to test stemness, upon targeting CAF-specific Tie2 activity by gene silencing or with small molecule inhibitor. Immunohistochemistry analysis was performed to locate the distribution of Tie2 and αSMA in primary tumors of oral carcinoma. Prognosis in HNSCC patient cohort from The Cancer Genome Atlas (TCGA) study was analysed based on single sample gene set enrichment score (ssGSEA) and Kaplan-Meier analysis. RESULTS Autocrine or exogenous TGFβ-induction in CAF led to the recruitment of histone deacetylase 2 (HDAC2) on the promoter of Tie2-antagonist, Angiopoietin-2 (ANGPT2), resulting in its downregulation, leading to phosphorylation of Tie2 (Y992) and subsequent activation of SRC (Y418). This led to SRC/ROCK mediated αSMA-positive stress-fiber formation with gain of myofibroblast phenotype. The CAF-specific Tie2-signaling was responsible for producing embryonic-like cell state in co-cultured cancer cells; with enhanced tumor initiating ability. Tie2 activity in CAF exerted the dynamic gene expression reprogramming, with the upregulation of 'cell migration' and downregulation of 'protein biosynthesis' related gene-regulatory-network modules in malignant cells. The AUCell scores calculated for gene signatures derived from these modules showed significant concordance in independently reported scRNAseq studies of HNSCC tumors and significant association with poor prognosis in HNSCC patient cohort. CONCLUSIONS CAF-specific Tie2 activity may serve as direct stromal-target against cancer cell plasticity leading to poor prognosis of oral cancer patients. Overall, our work has provided wider applicability of Tie2-specific functions in tumor biology, along with its known role in endothelial cell-specific function.
Collapse
Affiliation(s)
- Paromita Mitra
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Uday Saha
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | | | - Priyanka Prasad
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
| | - Subhashree Jena
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Ankit Kumar Patel
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Umea University, Umea, Sweden
| | | | | | - Sillarine Kurkalang
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Comprehensive Cancer Center, University of Chicago Medicine, Chicago, IL, USA
| | - Sumitava Roy
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Arnab Ghosh
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Shantanu Saha Roy
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
| | | | - Nidhan Kumar Biswas
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Moulinath Acharya
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | | | | | | | - Arindam Maitra
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Sandeep Singh
- BRIC National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, WB, 741251, India.
- Regional Centre for Biotechnology, Faridabad, India.
| |
Collapse
|
48
|
Chen L, Li H, Liu J, Wang Y, Zhang S. Hollow Mesoporous Carbon Nanospheres Derived from Metal-Organic Frameworks for Efficient Sono-immunotherapy against Pancreatic Cancer. CYBORG AND BIONIC SYSTEMS 2025; 6:0247. [PMID: 40352815 PMCID: PMC12062583 DOI: 10.34133/cbsystems.0247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 01/20/2025] [Accepted: 03/01/2025] [Indexed: 05/14/2025] Open
Abstract
Sono-immunotherapy is expected to effectively enhance treatment efficacy and reduce mortality in patients with pancreatic cancer. Hence, efficient applicable sono-immunotherapy systems are urgently needed for the treatment of this condition. In this study, hollow mesoporous carbon (HMC) nanoparticles were prepared using the sacrificial template method. These nanoparticles had a porphyrin-like structure and could generate singlet oxygen more efficiently than commercial TiO2. Cellular assays showed that HMC killed tumor cells in the presence of ultrasonication, primarily by inducing apoptosis. HMC could also accelerate the release of immune factors by tumor cells, thereby activating dendritic cells and enhancing the efficacy of immunotherapy. Experiments in tumor-bearing mice and in situ pancreatic cancer tests showed that HMC, in combination with the small-molecule inhibitors of programmed cell death ligand 1, could reduce tumor growth via the generation of reactive oxygen species following ultrasonication. HMC could enhance the efficacy of immunotherapy by disrupting the immunosuppressive tumor microenvironment and promoting the accumulation of immune cells. Accordingly, in vivo sono-immunotherapy was achieved, and the growth of transplanted tumors and in situ tumors could be reduced. In conclusion, this study proposes a novel method for the preparation of HMC nanoparticles and demonstrates their potential in tumor treatment. Additionally, owing to their unique structure, these HMC nanoparticles could be used for different combination therapies tailored based on specific clinical requirements.
Collapse
Affiliation(s)
- Libin Chen
- Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology,
Cancer Hospital of China Medical University, Shenyang 110042, China
- Department of Ultrasound Medicine,
The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
| | - Haiwei Li
- Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology,
Cancer Hospital of China Medical University, Shenyang 110042, China
| | - Jing Liu
- Department of Radiology,
The First Hospital of China Medical University, Shenyang 110001, China
| | - Yunzhong Wang
- Department of Ultrasound Medicine,
The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
| | - Shengmin Zhang
- Department of Ultrasound Medicine,
The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
| |
Collapse
|
49
|
Glapiński F, Zając W, Fudalej M, Deptała A, Czerw A, Sygit K, Kozłowski R, Badowska-Kozakiewicz A. The Role of the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma: Recent Advancements and Emerging Therapeutic Strategies. Cancers (Basel) 2025; 17:1599. [PMID: 40427098 PMCID: PMC12110676 DOI: 10.3390/cancers17101599] [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: 04/07/2025] [Revised: 04/30/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic cancer (PC), with pancreatic ductal adenocarcinoma (PDAC) comprising about 90% of all cases, is one of the most aggressive and lethal solid tumors. PDAC remains one of the most significant challenges of oncology to this day due to its inadequate response to conventional treatment, gradual rise in incidence since 2004, and poor five-year survival rates. As cancer cells are the primary adversary in this uneven fight, they remain the primary research target. Nevertheless, increasing attention is being paid to the tumor microenvironment (TME). The most crucial TME constellation components are immune cells, especially macrophages, stellate cells and lymphocytes, fibroblasts, bacterial and fungal microflora, and neuronal cells. Depending on the particular phenotype of these cells, the composition of the microenvironment, and the cell ratio, patients can experience different disease outcomes and varying vulnerability to treatment approaches. This study aims to present the current knowledge and review the most up-to-date scientific findings regarding the microenvironment of PC. It contains detailed information on the structure and cellular composition of the stroma, including its impact on disease development, metastasis, and response to treatment, as well as the therapeutic opportunities that arise from targeting this tissue.
Collapse
Affiliation(s)
- Franciszek Glapiński
- Students’ Scientific Organization of Cancer Cell Biology, Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Weronika Zając
- Students’ Scientific Organization of Cancer Cell Biology, Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland
| | - Marta Fudalej
- Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.F.); (A.D.)
- Department of Oncology, National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
| | - Andrzej Deptała
- Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.F.); (A.D.)
| | - Aleksandra Czerw
- Department of Health Economics and Medical Law, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Department of Economic and System Analyses, National Institute of Public Health NIH—National Research Institute, 00-791 Warsaw, Poland
| | - Katarzyna Sygit
- Faculty of Health Sciences, Calisia University, 62-800 Kalisz, Poland
| | - Remigiusz Kozłowski
- Department of Management and Logistics in Healthcare, Medical University of Lodz, 90-131 Lodz, Poland
| | - Anna Badowska-Kozakiewicz
- Department of Oncological Propaedeutics, Medical University of Warsaw, 01-445 Warsaw, Poland; (M.F.); (A.D.)
| |
Collapse
|
50
|
Dong S, Chen X, Li X, Wang Y, Huang Q, Li Y, Jin J, Zhu X, Zhong Y, Cai Q, Xue C, Guo F, Huang L, Feng M, Liu B, Hu S. A conceptual exploration on the synergistic anti-tumor effects of high-order combination of OHSV2-DSTE FAP5/CD3, CAR-T cells, and immunotoxins in hepatocellular carcinoma. Front Immunol 2025; 16:1509087. [PMID: 40406146 PMCID: PMC12095149 DOI: 10.3389/fimmu.2025.1509087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Background Although the treatment landscape for advanced hepatocellular carcinoma (HCC) has seen significant advancements in the past decade with the introduction of immune checkpoint inhibitors and antiangiogenic drugs, progress has fallen short of expectations. Recently, a novel engineered oncolytic virus (OHSV2) that secretes dual-specific T-cell engagers (DSTEs) targeting the fibroblast activation protein (FAP) was developed and combined with GPC3-targeting CAR-T cells and immunotoxins to exert a synergistic antitumor effect. Methods OHSV2-DSTEFAP5/CD3 was initially generated by transducing the DSTEs engaging FAP5 on fibroblasts into the backbone of our oncolytic virus OHSV2. An innovative high-order combination was devised in a xenograft mouse model to conceptually explore whether enhanced anti-tumor effects could be achieved. Additionally, the underlying mechanisms of synergistic effects and safety profiles were preliminarily investigated. Results OHSV2-DSTEFAP5/CD3 effectively targeted and eliminated fibroblasts in vitro while maintaining cytotoxicity and inducing immune activation compared to parental OHSV2. In vivo, dose-adjusted combination therapy resulted in a remarkable antitumor effect compared to control treatments, leading to tumor regression in 40% of mice without significant toxicity to major organs. Mechanistically, rather than directly depleting fibroblasts, OHSV2-DSTEFAP5/CD3 played an essential role in priming T-cell proliferation, infiltration, and activation, and inhibiting the supportive interaction between cancer cells and fibroblasts. Conclusions This high-order combination represents a novel multiple-wave immunotherapeutic approach for HCC. Despite being a conceptual exploration, this strategy has demonstrated promising therapeutic efficacy and acceptable safety profiles.
Collapse
Affiliation(s)
- Shuang Dong
- Department of Medical Oncology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Chen
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei, China
| | - Xiaoyu Li
- Department of Medical Oncology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Wang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei, China
| | - Qing Huang
- Department of Medical Oncology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanxiang Li
- Department of Medical Oncology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Jin
- Wuhan Binhui Biopharmaceutical Co., Ltd, Wuhan, China
| | - Xianmin Zhu
- Department of Medical Oncology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Zhong
- Department of Medical Oncology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Cai
- Department of Medical Oncology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chang Xue
- Department of Medical Oncology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Guo
- Department of Pathology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Le Huang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Mingqian Feng
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Binlei Liu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei, China
- Wuhan Binhui Biopharmaceutical Co., Ltd, Wuhan, China
| | - Sheng Hu
- Department of Medical Oncology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|