1
|
Zhang P, Pan J, Lin S, Peng B, An C, Zhang J, Xu L, Lai Y, Yu H, Xu Z. Smart drug delivery platforms reprogramming cancer immune cycle to mitigate immune resistance of pancreatic tumors. Adv Drug Deliv Rev 2025; 224:115620. [DOI: 10.1016/j.addr.2025.115620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2025]
|
2
|
Bräutigam K, Skok K, Szymonski K, Rift CV, Karamitopoulou E. Tumor immune microenvironment in pancreatic ductal adenocarcinoma revisited - Exploring the "Space". Cancer Lett 2025; 622:217699. [PMID: 40204149 DOI: 10.1016/j.canlet.2025.217699] [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: 12/21/2024] [Revised: 03/24/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most deadly malignancies with a highly immunosuppressive tumor immune microenvironment (TIME) that hinders effective therapy. PDAC is characterized by significant heterogeneity in immune cell composition, spatial distribution and activation states, which impacts tumor progression and treatment response. Tumour-infiltrating lymphocytes (TILs), including CD4+ T-helper cells, CD8+ cytotoxic T-cells and FOXP3+ regulatory T-cells, play a key role in immune regulation, yet PDAC is largely an immunologically "cold" tumour with limited effector T-cell infiltration. The surrounding cellular microenvironment, particularly Cancer Associated Fibroblasts (CAFs) and macrophages, contributes to immune evasion by promoting a fibrotic and desmoplastic barrier that limits TIL infiltration. The prognostic significance of TILs is increasingly recognized, with higher densities correlating with improved survival, whereas regulatory T-cell infiltration and immunosuppressive stromal interactions are associated with poor outcomes. Emerging therapeutic strategies targeting the TIME (e.g., CAFs), immune checkpoint inhibitors, and TIL-based therapies offer the potential to overcome resistance. Future research must focus on optimizing immunotherapy strategies and unravelling the complex stromal-immune interactions to improve clinical translation.
Collapse
Affiliation(s)
- Konstantin Bräutigam
- Institute of Cancer Research, Centre for Evolution and Cancer, London, SM2 5NG, United Kingdom; Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland.
| | - Kristijan Skok
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria; Institute of Biomedical Sciences, Medical Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Krzysztof Szymonski
- Department of Pathomorphology, Jagiellonian University Medical College, Krakow, Poland
| | | | - Eva Karamitopoulou
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
3
|
Otieno MO, Powrózek T, Garcia-Foncillas J, Martinez-Useros J. The crosstalk within tumor microenvironment and exosomes in pancreatic cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189308. [PMID: 40180303 DOI: 10.1016/j.bbcan.2025.189308] [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: 10/01/2024] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/05/2025]
Abstract
Pancreatic cancer is one of the most malignant tumors with a grim prognosis. Patients develop chemoresistance that drastically decreases their survival. The chemoresistance is mainly attributed to deficient vascularization of the tumor, intratumoral heterogeneity and pathophysiological barrier due to the highly desmoplastic tumor microenvironment. The interactions of cells that constitute the tumor microenvironment change its architecture into a cancer-permissive environment and stimulate cancer development, metastasis and treatment response. The cell-cell communication in the tumor microenvironment is often mediated by exosomes that harbour a diverse repertoire of molecular cargo, such as proteins, lipids, and nucleic acid, including messenger RNAs, non-coding RNAs and DNA. Therefore, exosomes can serve as potential targets as biomarkers and improve the clinical management of pancreatic cancer to overcome chemoresistance. This review critically elucidates the role of exosomes in cell-cell communication within the tumor microenvironment and how these interactions can orchestrate chemoresistance.
Collapse
Affiliation(s)
- Michael Ochieng' Otieno
- Translational Oncology Division, OncoHealth Institute, Health Research Institute Fundación Jimenez Diaz, Fundación Jimenez Díaz University Hospital, Universidad Autonoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Tomasz Powrózek
- Department of Human Physiology of the Chair of Preclinical Sciences, Medical University in Lublin, 20-080 Lublin, Poland
| | - Jesus Garcia-Foncillas
- Translational Oncology Division, OncoHealth Institute, Health Research Institute Fundación Jimenez Diaz, Fundación Jimenez Díaz University Hospital, Universidad Autonoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; Medical Oncology Department, Fundación Jimenez Diaz University Hospital, 28040, Madrid, Spain
| | - Javier Martinez-Useros
- Translational Oncology Division, OncoHealth Institute, Health Research Institute Fundación Jimenez Diaz, Fundación Jimenez Díaz University Hospital, Universidad Autonoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; Area of Physiology, Department of Basic Health Sciences, Faculty of Health Sciences, Rey Juan Carlos Univer-Sity, 28922 Madrid, Spain.
| |
Collapse
|
4
|
Sugumar K, Alabd A, Alabd A, Hue JJ, Lyons J, Fields S, Wainberg Z, Zheng L, Coogle B, Kasi A, Grewal N, Kindler HL, Starr J, Sama AR, Winter JM. Exceptional responders to immunotherapy in pancreatic cancer: A multi-institutional case series of a rare occurrence. Oncotarget 2025; 16:427-442. [PMID: 40492845 DOI: 10.18632/oncotarget.28739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2025] Open
Abstract
INTRODUCTION Immunotherapy has emerged as a standard treatment option for multiple solid tumors. However, most patients with pancreatic cancer (PC) do not derive a significant benefit. Identification and analyses of exceptional responders could eventually offer hints as to why PC is resistant to immunotherapy. METHODS Oncologists from cancer centers in the United States were contacted to identify patients with PC who responded to immunotherapy. Exceptional responders were defined as those having either partial (PR) or complete response (CR) based on Response Evaluation Criteria in Solid Tumors, or biochemical response (CA 19-9 levels) after starting immunotherapy. Patients receiving concurrent chemotherapy were excluded. RESULTS 14 patients met inclusion criteria. Immunotherapy drugs included checkpoint inhibitors and macrophage inhibitors. Eight patients (42%) were MSI (microsatellite instability)-high. Radiologically, 82% had PR. Four patients (28%) had marked reduction in CA 19-9. The median progression-free survival was 12 months from the start of immunotherapy. Median survival was not reached. The 1- and 2-year survival probabilities were 80%, 70% respectively. CONCLUSION Majority of clinical trials evaluating immunotherapy in PC have yielded disappointing response rates compared to other solid tumors. Our case series adds to published data from early-phase trials supporting the promise of immunotherapy in some patients with PC.
Collapse
Affiliation(s)
- Kavin Sugumar
- Department of Surgery, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Andrew Alabd
- Department of Medicine, Cooper University Healthcare, Camden, NJ 08103, USA
| | - Andre Alabd
- Department of Urology, University of Indiana, Indianapolis, IN 46227, USA
| | - Jonathan J Hue
- Department of Surgery, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Josh Lyons
- Department of Surgery, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Sherri Fields
- Department of Medicine, UCLA/Santa Monica Cancer Center, CA 90404, USA
| | - Zev Wainberg
- Department of Medicine, UCLA/Santa Monica Cancer Center, CA 90404, USA
| | - Lei Zheng
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Brianna Coogle
- Department of Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Anup Kasi
- Department of Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Nicholas Grewal
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Hedy L Kindler
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Jason Starr
- Department of Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ashwin R Sama
- Department of Medicine, Jefferson Medical Oncology Associates, Philadelphia, PA 19107, USA
| | - Jordan M Winter
- Department of Surgery, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| |
Collapse
|
5
|
Heo SC, Nam IH, Keum BR, Yun YG, Lee JY, Kim HJ. C-X-C motif chemokine ligand 1 derived from oral squamous cell carcinoma promotes cancer-associated fibroblast differentiation and tumor growth. MOLECULAR BIOMEDICINE 2025; 6:40. [PMID: 40490643 DOI: 10.1186/s43556-025-00281-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 05/23/2025] [Accepted: 05/28/2025] [Indexed: 06/11/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs), the predominant stromal cells in the tumor microenvironment (TME), play a critical role in the progression of solid tumors, including oral squamous cell carcinoma (OSCC). However, the molecular mechanisms by which OSCC-derived factors mediate CAF differentiation remain incompletely understood. This study investigates the role of the C-X-C motif chemokine ligand 1 (CXCL1), secreted by OSCC cells, in promoting CAF differentiation and its downstream impact on tumor progression. Gingival fibroblasts (GFs) were treated with conditioned medium (CM) from various OSCC cell lines to assess their potential to induce CAF differentiation. Proteomic analysis using liquid chromatography-mass spectrometry identified CXCL1 as a key factor highly secreted in SCC25-derived CM, which exhibited the strongest capacity to induce CAF differentiation. CXCL1 synergistically enhanced TGF-β1-induced differentiation of GFs into α-smooth muscle actin (αSMA)- and vimentin-expressing CAFs by approximately 1.5-fold, confirming its co-stimulatory function. Conversely, silencing its receptor CXCR2 reduced CAF marker expression by over 50%, indicating a strong inhibitory effect on CAF differentiation. In vivo, co-injection of SCC25 cells with GFs significantly promoted tumor growth and stromal CAF marker expression, whereas CXCR2 knockdown in GFs led to a ~ 40% reduction in tumor volume and reduced αSMA/vimentin-positive CAFs. These findings establish CXCL1 as a pivotal mediator of CAF differentiation through CXCR2-dependent signaling, and highlight that the CXCL1-CXCR2 axis is a promising therapeutic target for modulating stromal-tumor interactions in OSCC.
Collapse
Affiliation(s)
- Soon Chul Heo
- Department of Oral Physiology, Periodontal Diseases Signaling Network Research Center, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Institute of Tissue Regeneration Engineering (ITREN), Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - In-Hye Nam
- Department of Oral and Maxillofacial Surgery, Dental Research Institute, and Dental and Life Science Institute, Pusan National University, School of Dentistry, Yangsan, 50612, Republic of Korea
| | - Bo Ram Keum
- Department of Oral Physiology, Periodontal Diseases Signaling Network Research Center, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Yeo Gyun Yun
- Institute of Tissue Regeneration Engineering (ITREN), Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jae-Yeol Lee
- Department of Oral and Maxillofacial Surgery, Dental Research Institute, and Dental and Life Science Institute, Pusan National University, School of Dentistry, Yangsan, 50612, Republic of Korea.
| | - Hyung Joon Kim
- Department of Oral Physiology, Periodontal Diseases Signaling Network Research Center, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
6
|
Ai H, Nie R, Wang X. Pathway Enrichment-Based Unsupervised Learning Identifies Novel Subtypes of Cancer-Associated Fibroblasts in Pancreatic Ductal Adenocarcinoma. Interdiscip Sci 2025; 17:477-495. [PMID: 40272703 DOI: 10.1007/s12539-025-00705-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 03/21/2025] [Accepted: 03/23/2025] [Indexed: 05/28/2025]
Abstract
Existing single-cell clustering methods are based on gene expressions that are susceptible to dropout events in single-cell RNA sequencing (scRNA-seq) data. To overcome this limitation, we proposed a pathway-based clustering method for single cells (scPathClus). scPathClus first transforms the single-cell gene expression matrix into a pathway enrichment matrix and generates its latent feature matrix. Based on the latent feature matrix, scPathClus clusters single cells using the method of community detection. Applying scPathClus to pancreatic ductal adenocarcinoma (PDAC) scRNA-seq datasets, we identified two types of cancer-associated fibroblasts (CAFs), termed csCAFs and gapCAFs, which highly expressed complement system and gap junction-related pathways, respectively. Spatial transcriptome analysis revealed that gapCAFs and csCAFs are located at cancer and non-cancer regions, respectively. Pseudotime analysis suggested a potential differentiation trajectory from csCAFs to gapCAFs. Bulk transcriptome analysis showed that gapCAFs-enriched tumors are more endowed with tumor-promoting characteristics and worse clinical outcomes, while csCAFs-enriched tumors confront stronger antitumor immune responses. Compared to established CAF subtyping methods, this method displays better prognostic relevance.
Collapse
Affiliation(s)
- Hongjing Ai
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Intelligent Pharmacy Interdisciplinary Research Center, China Pharmaceutical University, Nanjing, 211198, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Rongfang Nie
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Intelligent Pharmacy Interdisciplinary Research Center, China Pharmaceutical University, Nanjing, 211198, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Intelligent Pharmacy Interdisciplinary Research Center, China Pharmaceutical University, Nanjing, 211198, China.
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
7
|
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
|
8
|
Gupta T, Murtaza M. Advancing targeted therapies in pancreatic cancer: Leveraging molecular abberrations for therapeutic success. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:19-32. [PMID: 39988056 DOI: 10.1016/j.pbiomolbio.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 02/03/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Pancreatic cancer is one of the most deadly with poor prognosis and overall survival rate due to the dense stroma in the tumors which often is challenging for the delivery of drug to penetrate deep inside the tumor bed and usually results in the progression of cancer. The conventional treatment such as chemotherapy, radiotherapy or surgery shows a minimal benefit in the survival due to the drug resistance, poor penetration, less radiosensitivity or recurrence of tumor. There is an urgent demand to develop molecular-level targeted therapies to achieve therapeutic efficacy in the pancreatic ductal adenocarcinoma (PDAC) patients. The precision oncology focuses on the unique attributes of the patient such as epigenome, proteome, genome, microbiome, lifestyle and diet habits which contributes to promote oncogenesis. The targeted therapy helps to target the mutated proteins responsible for controlling growth, division and metastasis of tumor in the cancer cells. It is very important to consider all the attributes of the patient to provide the suitable personalized treatment to avoid any severe side effects. In this review, we have laid emphasis on the precision medicine; the utmost priority is to improve the survival of cancer patients by targeting molecular mutations through transmembrane proteins, inhibitors, signaling pathways, immunotherapy, gene therapy or the use of nanocarriers for the delivery at the tumor site. It will become beneficial therapeutic window to be considered for the advanced stage pancreatic cancer patients to prolong their survival rate.
Collapse
Affiliation(s)
- Tanvi Gupta
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan.
| | - Mohd Murtaza
- Fermentation & Microbial Biotechnology Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180016, India.
| |
Collapse
|
9
|
Sigirli S, Karakas D. Fibrotic Fortresses and Therapeutic Frontiers: Pancreatic Stellate Cells and the Extracellular Matrix in Pancreatic Cancer. Cancer Med 2025; 14:e70788. [PMID: 40437741 PMCID: PMC12119906 DOI: 10.1002/cam4.70788] [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: 12/26/2024] [Revised: 02/19/2025] [Accepted: 03/08/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is characterized by a unique tumor microenvironment (TME) that plays pivotal roles in cancer progression, angiogenesis, metastasis, and drug resistance. This complex and dynamic ecosystem comprises cancer cells, stromal cells, and extracellular matrix (ECM) components, which interact synergistically to drive cancer aggressiveness. Among the stromal cells, cancer-associated fibroblasts (CAFs) and pancreatic stellate cells (PSCs), mainly accepted as a group of CAFs, are central players in shaping the desmoplastic, hypoxic, and immunosuppressive stroma of PDAC. PSCs, the most abundant stromal cells in PDAC, are resident pancreatic cells that undergo phenotypic changes upon activation, driving tumor progression through the secretion of cytokines, growth factors, ECM components (e.g., collagen, hyaluronic acid, fibronectin), and matrix metalloproteinases. In addition to cellular elements, ECM components significantly contribute to cancer aggressiveness by forming a physical barrier that hinders drug penetration, activating signaling pathways through specific receptor interactions, and generating peptides originating from the fragmentation of proteins to induce cancer migration. Regarding their critical roles in tumor progression, therapeutic approaches targeting PSCs and the ECM have garnered increasing interest in recent years. However, PSCs and stromal components may exhibit dual roles, with the potential to both promote and suppress tumor progression under different conditions. Therefore, targeting PSCs or stroma may lead to unintended outcomes, including exacerbation of cancer aggressiveness. METHODS This review focuses on the multifaceted roles of PSCs in PDAC, particularly their interactions with cancer cells and their contributions to therapy resistance. Additionally, we discuss current and emerging therapeutic strategies targeting PSCs and the ECM components, including both preclinical and clinical efforts. CONCLUSION By synthesizing insights from recent literature, this review provides a comprehensive understanding of the role of PSCs in PDAC pathobiology and highlights potential therapeutic approaches targeting PSCs or ECM components to improve patient outcomes.
Collapse
Affiliation(s)
- Sila Sigirli
- Medical Biotechnology, Graduate School of HealthAcibadem Mehmet Ali Aydinlar UniversityIstanbulTurkiye
| | - Didem Karakas
- Medical Biotechnology, Graduate School of HealthAcibadem Mehmet Ali Aydinlar UniversityIstanbulTurkiye
| |
Collapse
|
10
|
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
|
11
|
Yan B, Fritsche AK, Haußner E, Inamdar TV, Laumen H, Boettcher M, Gericke M, Michl P, Rosendahl J. From Genes to Environment: Elucidating Pancreatic Carcinogenesis Through Genetically Engineered and Risk Factor-Integrated Mouse Models. Cancers (Basel) 2025; 17:1676. [PMID: 40427173 PMCID: PMC12110317 DOI: 10.3390/cancers17101676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic cancer is characterized by late diagnosis, therapy resistance, and poor prognosis, necessitating the exploration of early carcinogenesis and prevention methods. Preclinical mouse models have evolved from cell line-based to human tumor tissue- or organoid-derived xenografts, now to humanized mouse models and genetically engineered mouse models (GEMMs). GEMMs, primarily driven by oncogenic Kras mutations and tumor suppressor gene alterations, offer a realistic platform for investigating pancreatic cancer initiation, progression, and metastasis. The incorporation of inducible somatic mutations and CRISPR-Cas9 screening methods has expanded their utility. To better recapitulate tumor initiation triggered by inflammatory cues, common pancreatic risk factors are being integrated into model designs. This approach aims to decipher the role of environmental factors as secondary or parallel triggers of tumor initiation alongside oncogenic burdens. Emerging models exploring pancreatitis, obesity, diabetes, and other risk factors offer significant translational potential. This review describes current mouse models for studying pancreatic carcinogenesis, their combination with inflammatory factors, and their utility in evaluating pathogenesis, providing guidance for selecting the most suitable models for pancreatic cancer research.
Collapse
Affiliation(s)
- Bin Yan
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Anne-Kristin Fritsche
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany;
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany;
| | - Erik Haußner
- Institute of Molecular Medicine, Section for Molecular Medicine of Signal Transduction, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (E.H.); (M.B.)
| | - Tanvi Vikrant Inamdar
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| | - Helmut Laumen
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| | - Michael Boettcher
- Institute of Molecular Medicine, Section for Molecular Medicine of Signal Transduction, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (E.H.); (M.B.)
| | - Martin Gericke
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany;
| | - Patrick Michl
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| |
Collapse
|
12
|
Roscigno G, Jacobs S, Toledo B, Borea R, Russo G, Pepe F, Serrano MJ, Calabrò V, Troncone G, Giovannoni R, Giovannetti E, Malapelle U. The potential application of stroma modulation in targeting tumor cells: focus on pancreatic cancer and breast cancer models. Semin Cancer Biol 2025:S1044-579X(25)00060-4. [PMID: 40373890 DOI: 10.1016/j.semcancer.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer development and spreading being considered as "the dark side of the tumor". Within this term tumor cells, immune components, supporting cells, extracellular matrix and a myriad of bioactive molecules that synergistically promote tumor development and therapeutic resistance, are included. Recent findings revealed the profound impacts of TME on cancer development, serving as physical support, critical mediator and biodynamic matrix in cancer evolution, immune modulation, and treatment outcomes. TME targeting strategies built on vasculature, immune checkpoints, and immuno-cell therapies, have paved the way for revolutionary clinical interventions. On this basis, the relevance of pre-clinical and clinical investigations has rapidly become fundamental for implementing novel therapeutical strategies breaking cell-cell and cell -mediators' interactions between TME components and tumor cells. This review summarizes the key players in the breast and pancreatic TME, elucidating the intricate interactions among cancer cells and their essential role for cancer progression and therapeutic resistance. Different tumors such breast and pancreatic cancer have both different and similar stroma features, that might affect therapeutic strategies. Therefore, this review aims to comprehensively evaluate recent findings for refining breast and pancreatic cancer therapies and improve patient prognoses by exploiting the TME's complexity in the next future.
Collapse
Affiliation(s)
- Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy.
| | - Sacha Jacobs
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| | - Belen Toledo
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain.
| | - Roberto Borea
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| | - Gianluca Russo
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Maria Jose Serrano
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy; GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception Group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain.
| | - Viola Calabrò
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Roberto Giovannoni
- Department of Biology, Genetic Unit, University of Pisa, Via Derna 1, 56126 Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy.
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| |
Collapse
|
13
|
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
|
14
|
Strippoli R. Colorectal cancer peritoneal metastasis is promoted by tissue-specific fibroblasts that can arise in response to various local disorders. Cancer Lett 2025; 625:217749. [PMID: 40320040 DOI: 10.1016/j.canlet.2025.217749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/09/2025]
Affiliation(s)
- Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy.
| |
Collapse
|
15
|
Seidel T, Ohri N, Glaß M, Sunami Y, Müller LP, Kleeff J. Stromal Cells in Early Inflammation-Related Pancreatic Carcinogenesis-Biology and Its Potential Role in Therapeutic Targeting. Cancers (Basel) 2025; 17:1541. [PMID: 40361466 DOI: 10.3390/cancers17091541] [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: 04/03/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
The stroma of healthy pancreases contains various non-hematopoietic, non-endothelial mesenchymal cells. It is altered by chronic inflammation which in turn is a major contributor to the development of pancreatic adenocarcinoma (PDAC). In PDAC, the stroma plays a decisive and well-investigated role for tumor progression and therapy response. This review addresses the central role of stromal cells in the early inflammation-driven development of PDAC. It focuses on major subpopulations of pancreatic mesenchymal cells, i.e., fibroblasts, pancreatic stellate cells, and multipotent stroma cells, particularly their activation and functional alterations upon chronic inflammation including the development of different types of carcinoma-associated fibroblasts. In the second part, the current knowledge on the impact of activated stroma cells on acinar-to-ductal metaplasia and the transition to pancreatic intraepithelial neoplasia is summarized. Finally, putative strategies to target stroma cells and their signaling in early pancreatic carcinogenesis are reflected. In summary, the current data show that the activation of pancreatic stroma cells and the resulting fibrotic changes has pro- and anti-carcinogenetic effects but, overall, creates a carcinogenesis-promoting microenvironment. However, this is a dynamic process and the therapeutic targeting of specific pathways and cells requires in-depth knowledge of the molecular interplay of various cell types.
Collapse
Affiliation(s)
- Tina Seidel
- Department of Internal Medicine, University Hospital Halle, 06120 Halle (Saale), Germany
| | - Nupur Ohri
- Department of Visceral, Vascular and Endocrine Surgery, University Hospital Halle, 06120 Halle (Saale), Germany
| | - Markus Glaß
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany
| | - Yoshiaki Sunami
- Department of Visceral, Vascular and Endocrine Surgery, University Hospital Halle, 06120 Halle (Saale), Germany
| | - Lutz P Müller
- Department of Internal Medicine, University Hospital Halle, 06120 Halle (Saale), Germany
| | - Jörg Kleeff
- Department of Visceral, Vascular and Endocrine Surgery, University Hospital Halle, 06120 Halle (Saale), Germany
| |
Collapse
|
16
|
Brewer G, Savage P, Fortier AM, Zhao H, Pacis A, Wang YC, Zuo D, de Nobrega M, Pedersen A, Cassel de Camps C, Souleimanova M, Ramos VM, Ragoussis J, Park M, Moraes C. Invasive phenotypes of triple-negative breast cancer-associated fibroblasts are mechanosensitive, AhR-dependent and correlate with disease state. Acta Biomater 2025:S1742-7061(25)00314-9. [PMID: 40318744 DOI: 10.1016/j.actbio.2025.04.061] [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: 11/26/2024] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Cancer associated fibroblasts (CAFs) play a critically important role in facilitating tumour cell invasion during metastasis. They also modulate local biophysical features of the tumour microenvironment through the formation of fibrotic foci, which have been correlated with breast cancer aggression. However, the impact of the evolving three-dimensional biophysical tumour microenvironment on CAF function remains undefined. Here, by isolating CAFs from primary human triple-negative breast cancer tissue at the time of surgery, we find that their ability to remodel the local microenvironment and invade into a three-dimensional matrix correlates with disease state. We then engineered culture models to systematically deconstruct and recreate mechanical tissue features of early breast cancer fibrotic foci; and demonstrate that invasion is mechanically-activated only in CAFs from patients with no detectable pre-existing metastases, but is independent of mechanical cues in CAFs isolated from patients with later-stage axillary lymph node metastases. By comparing the differential transcriptional response of these cells to microenvironmental tissue stiffness, we identify the aryl hydrocarbon receptor (AhR) as being significantly upregulated in invasive sub-populations of both mechanically-activated and mechanically-insensitive CAFs. Increasing AhR expression in CAFs induced invasion, while suppressing AhR significantly reduced invasion in both mechanically-activated and mechanically-insensitive CAF populations, even on stiffnesses that recapitulate late-stage disease. This work therefore uses mechanobiological analyses to identify AhR as a mediator of CAF invasion, providing a potential stratification marker to identify those patients who might respond to future mechanics-based prophylactic therapies, and provides a targetable mechanism to limit CAF-associated metastatic disease progression in triple-negative breast cancer patients. STATEMENT OF SIGNIFICANCE: By designing a mechanically-tunable tissue-engineered model of fibroblastic foci, and using this to culture patient-derived cancer-associated fibroblasts, we demonstrate that these cells are differentially mechanosensitive, depending on disease stage of the patient. While comparing transcriptomic profiles of patient-derived cells produces too many pathways to screen, identifying the pathways activated by local tissue mechanics that were common across each patient allowed us to identify a specific target to limit fibroblast invasion. This broad discovery strategy may be useful across a variety of biomaterials-based tissue engineered models; and these specific findings suggest (1) a strategy to identify patients who might respond to CAF- or matrix-targeting therapies, and (2) a specific actionable target to limit CAF-associated metastatic disease progression.
Collapse
Affiliation(s)
- Gabrielle Brewer
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada; Department of Biochemistry, McGill University, 3649 Promenade Sir-William-Osler, Montréal, QC, Canada
| | - Paul Savage
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada; Department of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montréal, QC, Canada
| | - Anne-Marie Fortier
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada
| | - Hong Zhao
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada
| | - Alain Pacis
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada
| | - Yu-Chang Wang
- Department of Human Genetics, McGill University, 3640 University, Montreal, QC; Genome Innovation Centre, 740 Dr Penfield Ave, Montreal, QC
| | - Dongmei Zuo
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada
| | - Monyse de Nobrega
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada; Department of Genetics and Molecular Biology, State University of Londrina, Londrina, PR, Brazil
| | - Annika Pedersen
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada; Department of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montréal, QC, Canada
| | - Camille Cassel de Camps
- Department of Biomedical Engineering, McGill University, 3775 rue University, Montréal, QC, Canada
| | - Margarita Souleimanova
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada
| | - Valentina Muñoz Ramos
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada
| | - Jiannis Ragoussis
- Department of Human Genetics, McGill University, 3640 University, Montreal, QC; Genome Innovation Centre, 740 Dr Penfield Ave, Montreal, QC
| | - Morag Park
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada; Department of Biochemistry, McGill University, 3649 Promenade Sir-William-Osler, Montréal, QC, Canada; Department of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montréal, QC, Canada; Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Montréal, QC, Canada.
| | - Christopher Moraes
- Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Avenues des Pins, Montréal, QC, Canada; Department of Biomedical Engineering, McGill University, 3775 rue University, Montréal, QC, Canada; Department of Chemical Engineering, McGill University, 3610 rue University, Montréal, QC, Canada.
| |
Collapse
|
17
|
Liu H, Dilger JP. Different strategies for cancer treatment: Targeting cancer cells or their neighbors? Chin J Cancer Res 2025; 37:289-292. [PMID: 40353083 PMCID: PMC12062981 DOI: 10.21147/j.issn.1000-9604.2025.02.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 04/10/2025] [Indexed: 05/14/2025] Open
Affiliation(s)
- Hengrui Liu
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1TN, UK
- Department of Oncology, University of Cambridge, Cambridge CB2 1TN, UK
| | - James P. Dilger
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
18
|
Finger AM, Hendley AM, Figueroa D, Gonzalez H, Weaver VM. Tissue mechanics in tumor heterogeneity and aggression. Trends Cancer 2025:S2405-8033(25)00096-2. [PMID: 40307158 DOI: 10.1016/j.trecan.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 03/10/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Tumorigenesis ensues within a heterogeneous tissue microenvironment that promotes malignant transformation, metastasis and treatment resistance. A major feature of the tumor microenvironment is the heterogeneous population of cancer-associated fibroblasts and myeloid cells that stiffen the extracellular matrix. The heterogeneously stiffened extracellular matrix in turn activates cellular mechanotransduction and creates a hypoxic and metabolically hostile microenvironment. The stiffened extracellular matrix and elevated mechanosignaling also drive tumor aggression by fostering tumor cell growth, survival, and invasion, compromising antitumor immunity, expanding cancer stem cell frequency, and increasing mutational burden, which promote intratumor heterogeneity. Delineating the molecular mechanisms whereby tissue mechanics regulate these phenotypes should help to clarify the basis for tumor heterogeneity and cancer aggression and identify novel therapeutic targets that could improve patient outcome. Here, we discuss the role of the extracellular matrix in driving cancer aggression through its impact on tumor heterogeneity.
Collapse
Affiliation(s)
- Anna-Marie Finger
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA 94143; Current address: Liver Disease Research, Global Drug Discovery, Novo Nordisk A/S, Malov, Denmark
| | - Audrey Marie Hendley
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA 94143
| | - Diego Figueroa
- Department of Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Hugo Gonzalez
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA 94143; Current address: Laboratory of Tumor Microenvironment and Metastasis, Centro Ciencia & Vida, Santiago, Chile
| | - Valerie Marie Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA 94143; Department of Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
19
|
Jing H, Gao Y, Jing L, Yang H, Liu S. Recent advances in therapeutic use of transforming growth factor-beta inhibitors in cancer and fibrosis. Front Oncol 2025; 15:1489701. [PMID: 40352593 PMCID: PMC12061708 DOI: 10.3389/fonc.2025.1489701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 04/03/2025] [Indexed: 05/14/2025] Open
Abstract
Transforming growth factor-beta (TGF-β) has long been known to be associated with early embryonic development and organogenesis, immune supervision, and tissue repair and homeostasis in adults. TGF-β has complex roles in fibrosis and cancer that may be opposing at different stages of these diseases. Under pathological conditions, overexpression of TGF-β causes epithelial-mesenchymal transition, deposition of extracellular matrix, and formation of cancer-associated fibroblasts, leading to fibrotic disease or cancer. Fibroblasts, epithelial cells, and immune cells are the most common targets of TGF-β, while fibrosis and cancer are the most common TGF-β-associated diseases. Given the critical role of TGF-β and its downstream molecules in fibrosis and progression of cancer, therapies targeting TGF-β signaling appear to be a promising strategy. Preclinical and clinical studies have investigated therapies targeting TGF-β, including antisense oligonucleotides, monoclonal antibodies, and ligand traps. However, development of targeted TGF-β therapy has been hindered by systemic cytotoxicity. This review discusses the molecular mechanisms of TGF-β signaling and highlights targeted TGF-β therapy for cancer and fibrosis as a therapeutic strategy for related diseases.
Collapse
Affiliation(s)
- Hanhui Jing
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yan Gao
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Linyuan Jing
- Department of Integrated Chinese and Western Medicine, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Hanyu Yang
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shanglong Liu
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
20
|
Chen X, Sun S, Zhao J, Yu S, Chen J, Chen X. Tumor-stroma ratio combined with PD-L1 identifies pancreatic ductal adenocarcinoma patients at risk for lymph node metastases. Br J Cancer 2025:10.1038/s41416-025-03019-z. [PMID: 40246986 DOI: 10.1038/s41416-025-03019-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Pathological examination of lymph node metastasis (LNM) is crucial for treating pancreatic ductal adenocarcinoma (PDAC). Although the tumour stroma is correlated with prognosis in multiple solid tumors, its role in detecting LNM in PDAC is unclear. Thus, this study aimed to investigate the relationship of tumor-stroma ratio (TSR) with LNM, survival and mutational profile in PDAC. METHODS In this multicenter retrospective study, we examined molecular and clinicopathologic features of 737 PDAC patients from 5 independent cohorts, including surgically resected and endoscopic ultrasound fine-needle aspiration (EUS-FNA) biopsy specimens. TSR was evaluated on hematoxylin and eosin-stained slides and classified as stroma-low (<50% stroma) or stroma-high (≥50% stroma). RESULTS Compared to TSR-high cases, TSR-low cases were significantly associated with LNM (P < 0.001). TSR could accurately distinguish patients with and without LNM with an area under curve (AUC) of 0.749, with the sensitivity and specificity of 76.5% and 71.6%, respectively. This accuracy of TSR for identifying LNM was further increased by adding other factors including PD-L1 expression or pretreatment serum CA19-9 levels. TSR showed similar levels of accuracy in analysis of resected tumor specimens and EUS-FNA biopsies. Moreover, we found that TSR could also identify residual nodal involvement after neoadjuvant therapy (NAT) using pretreatment EUS-FNA biopsy samples. Heterogeneous genetic alterations were observed between TSR-low and TSR-high subgroups. TSR was identified as an independent predictor of LNM and worse disease-free survival. Major findings were all reproducible in validation, EUS-FNA biopsy, and pre-treatment NAT EUS-FNA biopsy cohorts. CONCLUSIONS TSR served as a robust and reproducible biomarker that identifies patients at risk for LNM. TSR might be used to select treatment and management strategies for PDAC patients.
Collapse
Affiliation(s)
- Xianlong Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shanyue Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jiapeng Zhao
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuangni Yu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Jie Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Xinyuan Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
21
|
FENG LIWEN, CHEN YUTING, JIN WENYI. Research progress on cancer-associated fibroblasts in osteosarcoma. Oncol Res 2025; 33:1091-1103. [PMID: 40296919 PMCID: PMC12033999 DOI: 10.32604/or.2024.054207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/26/2024] [Indexed: 04/30/2025] Open
Abstract
Osteosarcoma (OS) is a prevalent primary bone malignancy with limited treatment options. Therefore, it is imperative to investigate and understand the mechanisms underlying OS pathogenesis. Cancer-associated fibroblasts (CAFs) are markedly abundant in tumor stromal cells and are essentially involved in the modulation of tumor occurrence and development. In recent years, CAFs have become a hotspot as researchers aim to elucidate CAF mechanisms that regulate tumor progression. However, most studies on CAFs are limited to a few common cancers, and their association with OS remains elusive. This review describes the role and current knowledge of CAFs in OS, focusing on their potential cellular origin, classification, and diverse functionality. It was found that CAFs influenced OS tumor cell signaling, proliferation, invasion, metastasis, epithelial-mesenchymal transition, stemness maintenance, angiogenesis, and the ability to modify immune system components. Furthermore, findings on other common cancers indicated that effective therapeutic strategies included the manipulation of CAF activation, targeting CAF-derived components, and depletion of CAFs by biomarkers. This review provides new insights and a theoretical basis for OS research.
Collapse
Affiliation(s)
- LIWEN FENG
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - YUTING CHEN
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - WENYI JIN
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430022, China
| |
Collapse
|
22
|
Mohammad SI, Vasudevan A, Hussein Alzewmel A, Rab SO, Ballal S, Kalia R, Bethanney Janney J, Ray S, Joshi KK, Yasin HA. The mutual effects of stearoyl-CoA desaturase and cancer-associated fibroblasts: A focus on cancer biology. Exp Cell Res 2025; 447:114508. [PMID: 40122505 DOI: 10.1016/j.yexcr.2025.114508] [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: 01/25/2025] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
The tumor microenvironment (TME) 's primary constituents that promote cancer development are cancer-associated fibroblasts (CAFs). Metabolic remodeling has been shown to control CAF activity, particularly aberrant lipid metabolism. SCD1 can be thought of as the primary enzyme controlling the fluidity of lipid bilayers by gradually converting saturated fatty acids into monounsaturated fatty acids. Furthermore, its crucial function in the onset and spread of cancer is well acknowledged. Even with the increasing amount of research on changes in lipid metabolism, this problem remains a relatively understudied aspect of cancer research. Blocking several fatty acid synthesis-related enzymes highly expressed in cancerous cells inhibits cell division and encourages apoptosis. This is the situation with SCD1, whose overexpression has been linked to several changed tumors and cells. Both genetic and pharmacological silencing of SCD1 in cancer cells prevents glucose-mediated lipogenesis and tumor cell growth. However, its role in CAFs, hence, cancer biology, has been less studied. This study aimed to review the role of SCD1 in CAF biology, shedding light on their function in cancer cell biology.
Collapse
Affiliation(s)
- Suleiman Ibrahim Mohammad
- Research Follower, INTI International University, 71800 Negeri Sembilan, Malaysia; Electronic Marketing and Social Media, Economic and Administrative Sciences, Zarqa University, Jordan.
| | - Asokan Vasudevan
- Faculty of Business and Communications, INTI International University, 71800, Negeri Sembilan, Malaysia.
| | - Ahmad Hussein Alzewmel
- 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
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Rishiv Kalia
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - J Bethanney Janney
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India; Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Hatif Abdulrazaq Yasin
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq
| |
Collapse
|
23
|
Vinaixa J, Martínez-Bosch N, Gibert J, Manero-Rupérez N, Santofimia-Castaño P, Baudou FG, Vera RE, Pease DR, Iglesias M, Sen S, Wang X, Almada LL, Marks DL, Moreno M, Iovanna JL, Rabinovich GA, Fernandez-Zapico ME, Navarro P. Nuclear Galectin-1 promotes KRAS-dependent activation of pancreatic cancer stellate cells. Proc Natl Acad Sci U S A 2025; 122:e2424051122. [PMID: 40172967 PMCID: PMC12002210 DOI: 10.1073/pnas.2424051122] [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: 11/19/2024] [Accepted: 02/28/2025] [Indexed: 04/04/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancers, primarily due to its complex tumor microenvironment (TME), which drives both disease progression and therapy resistance. Understanding the molecular mechanisms governing TME dynamics is essential for developing new treatment strategies for this devastating disease. In this study, we uncover an oncogenic role for Galectin-1 (Gal1), a glycan-binding protein abundantly expressed by activated pancreatic stellate cells (PSCs), a key component of the PDAC TME that orchestrates tumor progression. Our findings reveal that Gal1 expression is elevated in the nucleus of human PSCs in both tissue samples and cultured cell lines. Using chromatin immunoprecipitation followed by sequencing analysis (ChIP-seq), we identify Gal1 occupancy at the promoters of several cancer-associated genes, including KRAS, a pivotal oncogene involved in PDAC pathogenesis. We demonstrate that Gal1 binds to the KRAS promoter, sustaining KRAS expression in PSCs, which, in turn, maintains PSC activation and promotes the secretion of protumorigenic cytokines. Mechanistically, Gal1 is required to preserve histone H3 lysine 4 monomethylation levels and to recruit the histone methyltransferase MLL1 to target promoters. Collectively, our findings define a nuclear function of Gal1 in modulating the transcriptional landscape of cancer-associated genes in PSCs within the PDAC TME, mediated through an epigenetic mechanism. These insights enhance our understanding of PDAC pathology and open potential avenues for therapeutic interventions targeting intracellular Gal1.
Collapse
Affiliation(s)
- Judith Vinaixa
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
| | - Neus Martínez-Bosch
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
- Cancer Research Program, Hospital del Mar Research Institute, Associated Unit Hospital del Mar Research Institute/Institute of Biomedical Research of Barcelona-Spanish National Research Council (IIBB-CSIC), Barcelona08003, Spain
| | - Joan Gibert
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
| | - Noemí Manero-Rupérez
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
| | - Patricia Santofimia-Castaño
- Translational Research and Innovative Therapies Department, Cancer Research Center of Marseille, INSERM U1068, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, UMR 7258, Marseille13273, France
| | - Federico G. Baudou
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad de Buenos Aires1428, Argentina
- Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján6700, Provincia de Buenos Aires, Argentina
| | - Renzo E. Vera
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - David R. Pease
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - Mar Iglesias
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
- Departament of Pathology, Hospital del Mar, Barcelona08003, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid28029, Spain
| | - Sandhya Sen
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - Xiyin Wang
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - Luciana L. Almada
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - David L. Marks
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - Mireia Moreno
- Cancer Research Program, Hospital del Mar Research Institute, Barcelona08003, Spain
| | - Juan L. Iovanna
- Translational Research and Innovative Therapies Department, Cancer Research Center of Marseille, INSERM U1068, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, UMR 7258, Marseille13273, France
| | - Gabriel A. Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad de Buenos Aires1428, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires1428, Argentina
- Caixa Research Institute, Barcelona08022, Spain
| | - Martin E. Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN55905
| | - Pilar Navarro
- Cancer Research Program, Hospital del Mar Research Institute, Associated Unit Hospital del Mar Research Institute/Institute of Biomedical Research of Barcelona-Spanish National Research Council (IIBB-CSIC), Barcelona08003, Spain
- Department of Molecular and Cellular Biomedicine, Institute of Biomedical Research of Barcelona-Spanish National Research Council (IIBB-CSIC), Barcelona08036, Spain
- Institut d’Investigacions Biomediques August Pi Sunyer, Barcelona08036, Spain
| |
Collapse
|
24
|
Liu S, Liu C, He Y, Li J. Benign non-immune cells in tumor microenvironment. Front Immunol 2025; 16:1561577. [PMID: 40248695 PMCID: PMC12003390 DOI: 10.3389/fimmu.2025.1561577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/24/2025] [Indexed: 04/19/2025] Open
Abstract
The tumor microenvironment (TME) is a highly complex and continuous evolving ecosystem, consisting of a diverse array of cellular and non-cellular components. Among these, benign non-immune cells, including cancer-associated fibroblasts (CAFs), adipocytes, endothelial cells (ECs), pericytes (PCs), Schwann cells (SCs) and others, are crucial factors for tumor development. Benign non-immune cells within the TME interact with both tumor cells and immune cells. These interactions contribute to tumor progression through both direct contact and indirect communication. Numerous studies have highlighted the role that benign non-immune cells exert on tumor progression and potential tumor-promoting mechanisms via multiple signaling pathways and factors. However, these benign non-immune cells may play different roles across cancer types. Therefore, it is important to understand the potential roles of benign non-immune cells within the TME based on tumor heterogeneity. A deep understanding allows us to develop novel cancer therapies by targeting these cells. In this review, we will introduce several types of benign non-immune cells that exert on different cancer types according to tumor heterogeneity and their roles in the TME.
Collapse
Affiliation(s)
- Shaowen Liu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chunhui Liu
- The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Yuan He
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jun Li
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
25
|
Li YC, Zhang L, Wang YT, Hu H, Zhang ZY, Nie QQ, Zuo CJ. Role of EFNAs in Shaping the Tumor Immune Microenvironment and Their Impact on Pancreatic Adenocarcinoma Prognosis. Cancer Manag Res 2025; 17:693-712. [PMID: 40190415 PMCID: PMC11972607 DOI: 10.2147/cmar.s502401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
Purpose Due to the highly heterogeneous and immunosuppressed tumor microenvironment (TME), pancreatic adenocarcinoma (PAAD) has limited therapeutic options and an abysmal prognosis. Ephrin A 1-5 (EFNA1-5) have been shown to regulate tumorigenesis and metastasis in various cancers, but its role in PAAD remains unclear. Methods We comprehensively analyzed EFNA gene expression levels in pan-cancer and PAAD using the GEPIA and HPA databases. Then, we assessed the prognostic value of EFNA1-5 using the Kaplan-Meier plotter and nomogram model. Further exploration of the association of EFNA1-5 with clinicopathological features of PAAD used information from the UALCAN database, and the TIMER dataset was used to reveal the correlation between EFNA1-5 and the tumor immune microenvironment (TIME) of pancreatic cancer. In addition, cBioPortal Databases, GSEA, and GSCALite were used to explore gene changes, protein interactions, and biological functions. Finally, the oncogenic effect of EFNA5 was verified in vivo and in vitro. Results The expression levels of EFNA1-5 were significantly upregulated in PAAD. The expression of EFNA1/3/4/5 were significantly associated with overall survival (OS) and relapse-free survival (RFS) in PAAD patients. The high expression of EFNA2-5 were related to poor clinical features, such as higher tumor stage or grade and a wider range of lymph node metastasis. EFNA1-5 were closely associated with immune cell infiltration, CAFs, and MDSCs expression. Furthermore, EFNA5 is an independent risk factor for poor prognosis in PAAD patients, and it can promote the malignant progression of pancreatic cancer in vitro and in vivo. Conclusion Differential expression of EFNA1-5 is associated with TIME in pancreatic cancer, predicts different survival outcomes, and maybe a novel prognostic marker reflecting an immunosuppressive state and a potential therapeutic target.
Collapse
Affiliation(s)
- Yu-Chao Li
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Lu Zhang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Yi-Ting Wang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Hao Hu
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Ze-Yu Zhang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Qian-Qian Nie
- Department of Central Laboratory, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Chang-Jing Zuo
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
26
|
Lugovoi ME, Karshieva SS, Usatova VS, Voznyuk AA, Zakharova VA, Levin AA, Petrov SV, Senatov FS, Mironov VA, Belousov VV, Koudan EV. The design of the spheroids-based in vitro tumor model determines its biomimetic properties. BIOMATERIALS ADVANCES 2025; 169:214178. [PMID: 39799900 DOI: 10.1016/j.bioadv.2025.214178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 12/25/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Cancer, one of the world's deadliest diseases, is expected to claim an estimated 16 million lives by 2040. Three-dimensional (3D) models of cancer have become invaluable tools for the study of tumor biology and the development of new therapies. The tumor microenvironment (TME) is a determinant of tumor progression and has implications for clinical therapies. Cancer-associated fibroblasts (CAFs) are one of the most important components of the TME. Modeling the interactions between cancer cells and CAFs in vitro can help to create biomimetic tumor equivalents for elucidating the causes of cancer growth and assessing the effectiveness of therapies. Here, we are investigated the effect of the mutual arrangement of tumor cells and fibroblasts on the formation of tumor models and their biomimetic properties. Pancreatic tumor models of three different designs were formed by the bioprinting method. Gelatin-alginate hydrogels with and without PANC-1 (pancreatic cancer) and NIH/3 T3 (mouse fibroblasts) cells, as well as their homo- and heterospheroids, were used as bioink. To enable bioprinting, we have chosen the most suitable compositions of alginate and gelatin that provide both good printability and cell proliferation activity. We also have investigated the kinetics of spheroid formation to identify the optimal cultivation parameters for achieving spheroid sizes suitable for bioprinting. All tumor models remained viable for 3-4 weeks. At the same time, the patterns of model development in the cultivation process and the biomimetic properties of the final tissue-engineered structures depended on the model design.
Collapse
Affiliation(s)
- Maksim E Lugovoi
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Saida Sh Karshieva
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Veronika S Usatova
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117997 Moscow, Russia
| | - Amina A Voznyuk
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Vasilina A Zakharova
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Aleksandr A Levin
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Stanislav V Petrov
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Fedor S Senatov
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Vladimir A Mironov
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia
| | - Vsevolod V Belousov
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117997 Moscow, Russia
| | - Elizaveta V Koudan
- National University of Science and Technology MISIS, 119049, Leninskiy pr. 4, Moscow, Russia.
| |
Collapse
|
27
|
Quemerais C, Jean C, Brunel A, Decaup E, Labrousse G, Audureau H, Raffenne J, Belhabib I, Cros J, Perraud A, Dusetti N, Nicolle R, Mathonnet M, Pyronnet S, Martineau Y, Fanjul M, Bousquet C. Unveiling FKBP7 as an early endoplasmic reticulum sentinel in pancreatic stellate cell activation, collagen remodeling and tumor progression. Cancer Lett 2025; 614:217538. [PMID: 39924075 DOI: 10.1016/j.canlet.2025.217538] [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: 12/09/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
In pancreatic ductal adenocarcinoma (PDAC), fibroblast activation leads to excessive secretion of extracellular matrix (ECM) and soluble factors that regulate tumor progression, prompting investigation into endoplasmic reticulum (ER)-resident proteins that may support this activation. We identified FKBP7, a peptidyl-prolyl isomerase in the ER, as overexpressed in PDAC stroma compared to cancer cells, and in patients with favorable prognosis. Analysis of single-cell RNA sequencing databases revealed FKBP7 expression in pancreatic stellate cells (PSCs) and cancer-associated fibroblasts (CAFs). When analyzed by immunohistochemistry on PDAC patient tissues, FKBP7 emerged as an early activation marker in the preneoplastic stroma, preceding αSMA expression, and responding to FAK- and TGFβ-induced stiffening and pro-fibrotic programs in PSCs. Functional analyses revealed that FKBP7 knockdown in PSCs enhanced contractility, Rho/FAK signaling, and secretion of pro-inflammatory cytokines as well as remodeling of type I collagen, promoting an activated phenotype and accelerating tumor growth in vivo. Conversely, FKBP7 expression supported a tumor-restraining (i.e. encapsulating) ECM characterized by type IV collagen. Mechanistically, FKBP7 interacts with BiP, and blocking this interaction instead leads to increased PSC secretion of type I collagen. Thus, FKBP7 serves as a novel PSC marker and ER regulator in a complex with BiP of the secretion of specific collagen subtypes, highlighting its potential to mediate ECM normalization and constrain PDAC tumorigenesis.
Collapse
Affiliation(s)
- Christophe Quemerais
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Christine Jean
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Alexia Brunel
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Emilie Decaup
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Guillaume Labrousse
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Hippolyte Audureau
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Jérôme Raffenne
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Ismahane Belhabib
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Jérôme Cros
- Department of Pathology, Beaujon-Bichat University Hospital - Paris Diderot University, Clichy, France
| | - Aurélie Perraud
- EA 3842 Laboratory, Medicine and Pharmacy Faculties, University of Limoges, France
| | - Nelson Dusetti
- Cancer Research Center of Marseille (CRCM), INSERM UMR-1068, CNRS UMR-7258, Marseille, France
| | - Remy Nicolle
- Center of Research on Inflammation (CRI), INSERM U1149, Paris, France
| | - Muriel Mathonnet
- EA 3842 Laboratory, Medicine and Pharmacy Faculties, University of Limoges, France
| | - Stéphane Pyronnet
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Yvan Martineau
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Marjorie Fanjul
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Corinne Bousquet
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France.
| |
Collapse
|
28
|
Maru SY, Wetzel M, Mitchell JT, Gross NE, Andaloori L, Howe K, Kartalia E, Mo G, Leatherman J, Ho WJ, Fertig EJ, Kagohara LT, Pearce EJ, Jaffee EM. Antigen-presenting cancer-associated fibroblasts in murine pancreatic tumors differentially control regulatory T cell phenotype and function via CXCL9 and CCL22. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645833. [PMID: 40236227 PMCID: PMC11996409 DOI: 10.1101/2025.03.27.645833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a complex tumor microenvironment (TME) including stromal cells that influence resistance to therapy. Recent studies have revealed that stromal cancer-associated fibroblasts (CAFs) are heterogeneous in origin, gene expression, and function. Antigen-presenting CAFs (apCAFs), are defined by major histocompatibility complex (MHC)-II expression and can activate effector CD4 + T cells that have the potential to contribute to the anti-cancer immune response, but also can induce regulatory T cell (Treg) differentiation. Whether apCAFs promote or restrain the antitumor response remains uncertain. Using tumor clones of the KPC murine PDAC model differing in sensitivity to immune checkpoint blockade (ICB), we found that immunosensitive (sKPC) tumors were characterized by higher immune cell and apCAF infiltration than resistant (rKPC) tumors. IMC analysis showed proximity of apCAFs and CD4 + T cells in both sKPC and rKPC tumors implicating interaction within the TME. apCAF-depleted sKPC tumor-bearing mice had diminished sensitivity to ICB. apCAFs from both sKPC and rKPC tumors activated tumor-infiltrating CD4 + T cells and induced Treg differentiation. However, transcriptomic analysis showed that Tregs induced by apCAFs were overexpressed for immunosuppressive genes in rKPCs relative to sKPCs, and that this is associated with differential chemokine signaling from apCAFs depending on tumor origin. Together these data implicate apCAFs as important mediators of the antitumor immune response, modulation of which could facilitate the development of more effective anti-tumor immune based approaches for PDAC patients.
Collapse
|
29
|
Ganbold M, Louphrasitthiphol P, Miyamoto T, Miyazaki Y, Oda T, Tominaga K, Isoda H. Isorhamnetin exerts anti-proliferative effect on cancer-associated fibroblasts by inducing cell cycle arrest. Biomed Pharmacother 2025; 185:117954. [PMID: 40031374 DOI: 10.1016/j.biopha.2025.117954] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/16/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025] Open
Abstract
Isorhamnetin (ISO), a dietary flavonoid, has been shown to possess antioxidant, anti-cancer, and anti-inflammatory properties. Cancer-associated fibroblasts (CAFs), found in the tumor microenvironment of several types of cancer including pancreatic ductal adenocarcinoma (PDAC) impact the tumor growth and development of chemoresistance. Thus, modulating CAFs is an attractive mean to increase the efficacy of therapies targeting cancer cells. In this study, the anti-proliferative effect of ISO and the underlying transcriptomic profile of ISO-treated PDAC-derived CAFs were investigated. ISO treatment showed a time- and concentration-dependent decrease in cell viability with a slight increase in apoptotic cells. Microarray and cell cycle analyses revealed ISO induced downregulation of pathways in cell cycle and DNA replication; and G2/M checkpoint. Cell cycle analysis showed cells in the G2/M phase were increased. In response to the treatment, hallmark for p53 pathway genes, known to regulate cell cycle checkpoints, were highly upregulated. Moreover, ISO-treated cells had an increased area of the mitochondrial network, but lower mitochondrial membrane potential accompanied by a decrease of ATP production, measured by oxygen consumption rate. Inflammatory gene expression of IL1A1, IL6, CXCL1, and LIF were significantly inhibited in ISO-treated CAFs. Taken together, our results demonstrated that the cytostatic effect of ISO on human CAFs was mediated by inducing cell cycle arrest at G2/M phase associated with activation of p21, impaired mitochondrial homeostasis, and inhibition of inflammatory mediators gene expression, warranting further investigation for its use in combinatorial therapy that target both the cancer and the tumor microenvironment as a whole.
Collapse
Affiliation(s)
- Munkhzul Ganbold
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Science and Technology (AIST), Tsukuba, Japan
| | - Pakavarin Louphrasitthiphol
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan; Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford, UK
| | - Takafumi Miyamoto
- Department of Internal Medicine (Endocrinology and Metabolism), Institute of Medicine, University of Tsukuba, Tsukuba, Japan; Transborder Medical Research Center, University of Tsukuba, Tsukuba, Japan
| | - Yoshihiro Miyazaki
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tatsuya Oda
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kenichi Tominaga
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Science and Technology (AIST), Tsukuba, Japan
| | - Hiroko Isoda
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Science and Technology (AIST), Tsukuba, Japan; Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan; Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
30
|
Jia H, Chen X, Zhang L, Chen M. Cancer associated fibroblasts in cancer development and therapy. J Hematol Oncol 2025; 18:36. [PMID: 40156055 PMCID: PMC11954198 DOI: 10.1186/s13045-025-01688-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/12/2025] [Indexed: 04/01/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are key players in cancer development and therapy, and they exhibit multifaceted roles in the tumor microenvironment (TME). From their diverse cellular origins, CAFs undergo phenotypic and functional transformation upon interacting with tumor cells and their presence can adversely influence treatment outcomes and the severity of the cancer. Emerging evidence from single-cell RNA sequencing (scRNA-seq) studies have highlighted the heterogeneity and plasticity of CAFs, with subtypes identifiable through distinct gene expression profiles and functional properties. CAFs influence cancer development through multiple mechanisms, including regulation of extracellular matrix (ECM) remodeling, direct promotion of tumor growth through provision of metabolic support, promoting epithelial-mesenchymal transition (EMT) to enhance cancer invasiveness and growth, as well as stimulating cancer stem cell properties within the tumor. Moreover, CAFs can induce an immunosuppressive TME and contribute to therapeutic resistance. In this review, we summarize the fundamental knowledge and recent advances regarding CAFs, focusing on their sophisticated roles in cancer development and potential as therapeutic targets. We discuss various strategies to target CAFs, including ECM modulation, direct elimination, interruption of CAF-TME crosstalk, and CAF normalization, as approaches to developing more effective treatments. An improved understanding of the complex interplay between CAFs and TME is crucial for developing new and effective targeted therapies for cancer.
Collapse
Affiliation(s)
- Hongyuan Jia
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Xingmin Chen
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Linling Zhang
- Department of Respiratory and Critical Care, Chengdu Third People's Hospital, Chengdu, China
| | - Meihua Chen
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
31
|
Tan X, Rosin M, Appinger S, Deierl JC, Reichel K, Coolsen M, Valkenburg-van Iersel L, de Vos-Geelen J, de Jong EJM, Bednarsch J, Grootkoerkamp B, Doukas M, van Eijck C, Luedde T, Dahl E, Kather JN, Sivakumar S, Knoefel WT, Wiltberger G, Neumann UP, Heij LR. Stroma and lymphocytes identified by deep learning are independent predictors for survival in pancreatic cancer. Sci Rep 2025; 15:9415. [PMID: 40108402 PMCID: PMC11923104 DOI: 10.1038/s41598-025-94362-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: 07/16/2024] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers known to humans. However, not all patients fare equally poor survival, and a minority of patients even survives advanced disease for months or years. Thus, there is a clinical need to search corresponding prognostic biomarkers which forecast survival on an individual basis. To dig more information and identify potential biomarkers from PDAC pathological slides, we trained a deep learning (DL) model based U-net-shaped backbone. This DL model can automatically detect tumor, stroma and lymphocytes on whole slide images (WSIs) of PDAC patients. We performed an analysis of 800 PDAC scans, categorizing stroma in percentage (SIP) and lymphocytes in percentage (LIP) into two and three categories, respectively. The presented model achieved remarkable accuracy results with a total accuracy of 94.72%, a mean intersection of union rate of 78.66%, and a mean dice coefficient of 87.74%. Survival analysis revealed that SIP-mediate and LIP-high groups correlated with enhanced median overall survival (OS) across all cohorts. These findings underscore the potential of SIP and LIP as prognostic biomarkers for PDAC and highlight the utility of DL as a tool for PDAC biomarkers detecting on WSIs.
Collapse
Affiliation(s)
- Xiuxiang Tan
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Mika Rosin
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Simone Appinger
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Julia Campello Deierl
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Konrad Reichel
- Department of General, Visceral and Transplantation Surgery, Universitats Klinikum Essen, Essen, Germany
| | - Mariëlle Coolsen
- Department of Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Liselot Valkenburg-van Iersel
- Department of Internal Medicine, Division of Medical Oncology, GROW, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Judith de Vos-Geelen
- Department of Internal Medicine, Division of Medical Oncology, GROW, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Evelien J M de Jong
- Department of Internal Medicine, Division of Medical Oncology, GROW, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Jan Bednarsch
- Department of General, Visceral and Transplantation Surgery, Universitats Klinikum Essen, Essen, Germany
| | - Bas Grootkoerkamp
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Michail Doukas
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Casper van Eijck
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Edgar Dahl
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Shivan Sivakumar
- Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Wolfram Trudo Knoefel
- Department of General, Visceral and Pediatric Surgery, Heinrich Heine University, Düsseldorf, Germany
| | - Georg Wiltberger
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Ulf Peter Neumann
- Department of General, Visceral and Transplantation Surgery, Universitats Klinikum Essen, Essen, Germany
- Department of Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Lara R Heij
- Department of General, Visceral and Transplantation Surgery, Universitats Klinikum Essen, Essen, Germany
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
- Institute of Pathology, University Hospital Essen, Essen, Germany
| |
Collapse
|
32
|
Baltanás FC, Kramer-Drauberg M, García-Navas R, Patrucco E, Petrini E, Arnhof H, Olarte-San Juan A, Rodríguez-Ramos P, Borrajo J, Calzada N, Castellano E, Mair B, Kostyrko K, Hofmann MH, Ambrogio C, Santos E. SOS1 inhibitor BI-3406 shows in vivo antitumor activity akin to genetic ablation and synergizes with a KRAS G12D inhibitor in KRAS LUAD. Proc Natl Acad Sci U S A 2025; 122:e2422943122. [PMID: 40073053 PMCID: PMC11929440 DOI: 10.1073/pnas.2422943122] [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: 11/07/2024] [Accepted: 01/24/2025] [Indexed: 03/14/2025] Open
Abstract
We evaluated the in vivo therapeutic efficacy and tolerability of BI-3406-mediated pharmacological inhibition of SOS1 in comparison to genetic ablation of this universal Ras-GEF in various KRAS-dependent experimental tumor settings. Contrary to the rapid lethality caused by SOS1 genetic ablation in SOS2KO mice, SOS1 pharmacological inhibition by its specific inhibitor BI-3406 did not significantly affect animal weight/viability nor cause noteworthy systemic toxicity. Allograft assays using different KRASmut cell lines showed that treatment with BI-3406 impaired RAS activation and RAS downstream signaling and decreased tumor burden and disease progression as a result of both tumor-intrinsic and -extrinsic therapeutic effects of the drug. Consistent with prior genetic evidence and the KRASmut allografts assays in immunocompromised mice, our analyses using an in vivo model of KRASG12D-driven lung adenocarcinoma (LUAD) in immunocompetent mice showed that single, systemic BI-3406 treatment impaired tumor growth and downmodulated protumorigenic components of the tumor microenvironment comparably to SOS1 genetic ablation or to treatment with the specific KRASG12D inhibitor MRTX1133. Furthermore, markedly stronger, synergistic antitumor effects were observed upon concomitant treatment with BI-3406 and MRTX1133 in the same in vivo LUAD mouse model. Our data confirm SOS1 as an actionable therapy target in RAS-dependent cancers and suggest that BI-3406 treatment may yield clinical benefit both as monotherapy or as a potential combination partner for multiple RAS-targeting strategies.
Collapse
Affiliation(s)
- Fernando C. Baltanás
- Laboratorio 1. Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Salamanca37007, Spain
- Instituto de Biomedicina de Sevilla (IBiS)/Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Sevilla and Departamento de Fisiología Medica y Biofísica, Universidad de Sevilla, Sevilla41013, Spain
| | - Maximilian Kramer-Drauberg
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino10126, Italy
| | - Rósula García-Navas
- Laboratorio 1. Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Salamanca37007, Spain
| | - Enrico Patrucco
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino10126, Italy
| | - Ettore Petrini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino10126, Italy
| | - Heribert Arnhof
- Boehringer Ingelheim Regional Center Vienna GmbH & Co KG, Vienna1120, Austria
| | - Andrea Olarte-San Juan
- Laboratorio 1. Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Salamanca37007, Spain
| | - Pablo Rodríguez-Ramos
- Laboratorio 1. Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Salamanca37007, Spain
| | - Javier Borrajo
- Departamento de Ciencias Biomédicas y del Diagnóstico, Universidad de Salamanca, Salamanca37007, Spain
| | - Nuria Calzada
- Laboratorio 1. Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Salamanca37007, Spain
| | - Esther Castellano
- Laboratorio 5. Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Salamanca37007, Spain
| | - Barbara Mair
- Boehringer Ingelheim Regional Center Vienna GmbH & Co KG, Vienna1120, Austria
| | - Kaja Kostyrko
- Boehringer Ingelheim Regional Center Vienna GmbH & Co KG, Vienna1120, Austria
| | - Marco H. Hofmann
- Boehringer Ingelheim Regional Center Vienna GmbH & Co KG, Vienna1120, Austria
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino10126, Italy
| | - Eugenio Santos
- Laboratorio 1. Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas-Universidad de Salamanca and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Salamanca37007, Spain
| |
Collapse
|
33
|
Angeli S, Neophytou C, Kalli M, Stylianopoulos T, Mpekris F. The mechanopathology of the tumor microenvironment: detection techniques, molecular mechanisms and therapeutic opportunities. Front Cell Dev Biol 2025; 13:1564626. [PMID: 40171226 PMCID: PMC11958720 DOI: 10.3389/fcell.2025.1564626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
The mechanical properties of the tumor microenvironment (TME) undergo significant changes during tumor growth, primarily driven by alterations in extracellular (ECM) stiffness and tumor viscoelasticity. These mechanical changes not only promote tumor progression but also hinder therapeutic efficacy by impairing drug delivery and activating mechanotransduction pathways that regulate crucial cellular processes such as migration, proliferation, and resistance to therapy. In this review, we examine the mechanisms through which tumor cells sense and transmit mechanical signals to maintain homeostasis in the biomechanically altered TME. We explore current computational modelling strategies for mechanotransduction pathways, highlighting the need for developing models that incorporate additional components of the mechanosignaling machinery. Furthermore, we review available methods for measuring the mechanical properties of tumors in clinical settings and strategies aiming at restoring the TME and blocking deregulated mechanotransduction pathways. Finally, we propose that proper characterization and a deeper understanding of the mechanical landscape of the TME, both at the tissue and cellular levels, are essential for developing therapeutic strategies that account for the influence of mechanical forces on treatment efficacy.
Collapse
Affiliation(s)
| | | | | | | | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
34
|
Chang C, Lin C. Click Hydrogels to Assess Stiffness-Induced Activation of Pancreatic Cancer-Associated Fibroblasts and Its Impact on Cancer Cell Spreading. Chembiochem 2025; 26:e202400955. [PMID: 39794304 PMCID: PMC11907380 DOI: 10.1002/cbic.202400955] [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: 11/22/2024] [Revised: 01/02/2025] [Accepted: 01/10/2025] [Indexed: 01/13/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is marked by significant desmoplastic reactions, or the accumulation of excessive extracellular matrices. PDAC stroma has abnormally high stiffness, which alters cancer cell behaviors and creates a barrier for effective drug delivery. Unfortunately, clinical trials using a combination of chemotherapy and matrix-degrading enzyme have led to disappointing results, as the degradation of stromal tissue likely accelerated the dissemination of cancer cells. High matrix stiffness has been shown to activate cancer-associated fibroblasts (CAFs), increasing their interaction with pancreatic cancer cells (PCCs) through promoting proliferation, migration, and resistance to chemotherapy. With the advance of biomaterials science and engineering, it is now possible to design chemically defined matrices to understand the role of stiffness in activating pancreatic CAFs and how this may alter cancer cell migration. Here, we developed a norbornene-based click hydrogel system with independently tunable stiffness and cell adhesive ligand to evaluate stiffness-induced activation of CAFs and migration of PCCs. Our results show that matrix stiffness did not alter matrix deposition from CAFs but affected nuclear localization of Yes-associated protein (YAP). Our results also verify the role of CAFs on promoting PCC migration and an elevated substrate stiffness further increased PCC motility.
Collapse
Affiliation(s)
- Chun‐Yi Chang
- Weldon School of Biomedical EngineeringPurdue University206 Martin Jischke Dr.West LafayetteIN 47907USA
| | - Chien‐Chi Lin
- Weldon School of Biomedical EngineeringPurdue University206 Martin Jischke Dr.West LafayetteIN 47907USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer CenterIndianapolisIN 46202USA
| |
Collapse
|
35
|
Wang Y, Lu J, Chong X, Wang C, Chen X, Peng Z, Gu Y, Wang Y, Wang X, Li J, Gong J, Qi C, Yuan J, Lu Z, Lu M, Zhou J, Cao Y, Chen Y, Zhang C, Hou Z, Kou H, Shen L, Zhang X. PD-1 antibody camrelizumab plus apatinib and SOX as first-line treatment in patients with AFP-producing gastric or gastro-esophageal junction adenocarcinoma (CAP 06): a multi-center, single-arm, phase 2 trial. Signal Transduct Target Ther 2025; 10:100. [PMID: 40082418 PMCID: PMC11906745 DOI: 10.1038/s41392-025-02193-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 02/18/2025] [Accepted: 02/25/2025] [Indexed: 03/16/2025] Open
Abstract
Alpha-fetoprotein-producing gastric or gastro-esophageal junction (AFP-G/GEJ) cancer, a rare gastric cancer subtype, exhibits increased angiogenesis and more immunosuppression than non-AFP-G/GEJ cancer. The potential benefits of anti-angiogenic agents and immunotherapy for this specific subtype remain unknown. This multi-center, single-arm, phase 2 trial (ClinicalTrials.gov NCT04609176) evaluated the antitumor activity, safety, and biomarkers of camrelizumab plus apatinib and S-1 and oxaliplatin (SOX), followed by maintenance treatment with camrelizumab plus apatinib, as a first-line treatment in patients with AFP-G/GEJ adenocarcinoma. Primary endpoint was the confirmed objective response rate (ORR) per RECIST v1.1 in the full analysis set. Secondary endpoints were disease control rate (DCR), progression-free survival (PFS), overall survival (OS), duration of response, time to response, and safety. Between December 4, 2020, and August 4, 2023, 36 patients were enrolled and treated. The trial met its primary endpoint with a confirmed ORR of 66.7% (95% CI: 49.0-81.4). The DCR was 88.9% (95% CI: 73.9-96.9). With a median follow-up of 11.7 months (range: 3.2-37.9), the median PFS reached 7.8 months (95% CI: 4.9-12.3) and the median OS reached 18.0 months (95% CI: 10.5-NR). No new safety concerns were identified. In exploratory analysis, patients with durable clinical benefit exhibited higher pre-treatment (PD-1+) CD8+ T cell densities and effective scores. First-line treatment with camrelizumab plus apatinib and SOX, followed by maintenance treatment with camrelizumab plus apatinib, is effective and safe in AFP-G/GEJ adenocarcinoma. Further studies are necessary to validate these findings.
Collapse
Affiliation(s)
- Yakun Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jialin Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaoyi Chong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chang Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Xiaofeng Chen
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi Peng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanhong Gu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yizhuo Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Xicheng Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jian Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jifang Gong
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Changsong Qi
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Early Drug Development Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiajia Yuan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhihao Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ming Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jun Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanshuo Cao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yang Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Cheng Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhiguo Hou
- Jiangsu Hengrui Pharmaceuticals Co. Ltd, Shanghai, China
| | - Hongyi Kou
- Jiangsu Hengrui Pharmaceuticals Co. Ltd, Shanghai, China
| | - Lin Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Xiaotian Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China.
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, China.
| |
Collapse
|
36
|
Turlej E, Domaradzka A, Radzka J, Drulis-Fajdasz D, Kulbacka J, Gizak A. Cross-Talk Between Cancer and Its Cellular Environment-A Role in Cancer Progression. Cells 2025; 14:403. [PMID: 40136652 PMCID: PMC11940884 DOI: 10.3390/cells14060403] [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: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
The tumor microenvironment is a dynamic and complex three-dimensional network comprising the extracellular matrix and diverse non-cancerous cells, including fibroblasts, adipocytes, endothelial cells and various immune cells (lymphocytes T and B, NK cells, dendritic cells, monocytes/macrophages, myeloid-derived suppressor cells, and innate lymphoid cells). A constantly and rapidly growing number of studies highlight the critical role of these cells in shaping cancer survival, metastatic potential and therapy resistance. This review provides a synthesis of current knowledge on the modulating role of the cellular microenvironment in cancer progression and response to treatment.
Collapse
Affiliation(s)
- Eliza Turlej
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Aleksandra Domaradzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Justyna Radzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Dominika Drulis-Fajdasz
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Julita Kulbacka
- Departament of Molecular and Cellular Biology, Faculty of Pharmacy, Wrocław Medical University, Borowska 211A, 50-556 Wrocław, Poland;
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Agnieszka Gizak
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| |
Collapse
|
37
|
Meguro S, Nakanishi M. Cellular senescence in the cancer microenvironment. J Biochem 2025; 177:171-176. [PMID: 39760850 DOI: 10.1093/jb/mvaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/25/2024] [Accepted: 12/07/2024] [Indexed: 01/07/2025] Open
Abstract
In this ageing society, the number of patients suffering from age-related diseases, including cancer, is increasing. Cellular senescence is a cell fate that involves permanent cell cycle arrest. Accumulated senescent cells in tissues over time present senescence-associated secretory phenotype (SASP) and make the inflammatory context, disturbing the tumour microenvironment. In particular, the effect of senescent cancer-associated fibroblasts on cancer progression has recently come under the spotlight. Although scientific evidence on the impact of cellular senescence on cancer is emerging, the association between cellular senescence and cancer is heterogeneous and the comprehensive mechanism is still not revealed. Recently, a therapy targeting senescent cells, senotherapeutics, has been reported to be effective against cancer in preclinical research and even clinical trials. With further research, the development of senotherapeutics as a novel cancer therapy is expected.
Collapse
Affiliation(s)
- Satoru Meguro
- Division of Cancer Cell Biology, Institute of Medical Science, University of Tokyo, Shirokanedai 4-6-1, Minato-ku, Tokyo 108-8639, Tokyo, Japan
- Department of Urology, Fukushima Medical University School of Medicine, Fukushima, 960-1247, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, Institute of Medical Science, University of Tokyo, Shirokanedai 4-6-1, Minato-ku, Tokyo 108-8639, Tokyo, Japan
| |
Collapse
|
38
|
Affὸ S, Sererols-Viñas L, Garcia-Vicién G, Cadamuro M, Chakraborty S, Sirica AE. Cancer-Associated Fibroblasts in Intrahepatic Cholangiocarcinoma: Insights into Origins, Heterogeneity, Lymphangiogenesis, and Peritoneal Metastasis. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:378-396. [PMID: 39117110 DOI: 10.1016/j.ajpath.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/11/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024]
Abstract
Intrahepatic cholangiocarcinoma (iCCA) denotes a rare, highly malignant, and heterogeneous class of primary liver adenocarcinomas exhibiting phenotypic characteristics of cholangiocyte differentiation. Among the distinctive pathological features of iCCA, one that differentiates the most common macroscopic subtype (eg, mass-forming type) of this hepatic tumor from conventional hepatocellular carcinoma is a prominent desmoplastic reaction manifested as a dense fibro-collagenous-enriched tumor stroma. Cancer-associated fibroblasts (CAFs) represent the most abundant mesenchymal cell type in the desmoplastic reaction. Although the protumor effects of CAFs in iCCA have been increasingly recognized, more recent cell lineage tracing studies, advanced single-cell RNA sequencing, and expanded biomarker analyses have provided new awareness into their ontogeny, as well as underscored their biological complexity as reflected by the presence of multiple subtypes. In addition, evidence supports CAFs' potential to display cancer-restrictive roles, including immunosuppression. However, CAFs also play important roles in facilitating metastasis, as exemplified by lymph node metastasis and peritoneal carcinomatosis, which are common in iCCA. Herein, the authors provide a timely appraisal of the origins and phenotypic and functional complexity of CAFs in iCCA, together with providing mechanistic insights into lymphangiogenesis and peritoneal metastasis relevant to this lethal human cancer.
Collapse
Affiliation(s)
- Silvia Affὸ
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Laura Sererols-Viñas
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Gemma Garcia-Vicién
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Sanjukta Chakraborty
- Department of Medical Physiology, School of Medicine, Texas A&M Health Science Center, Bryan, Texas
| | - Alphonse E Sirica
- Department of Pathology (Emeritus), Virginia Commonwealth University School of Medicine, Richmond, Virginia.
| |
Collapse
|
39
|
Huang S, Soto AM, Sonnenschein C. The end of the genetic paradigm of cancer. PLoS Biol 2025; 23:e3003052. [PMID: 40100793 DOI: 10.1371/journal.pbio.3003052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
Genome sequencing of cancer and normal tissues, alongside single-cell transcriptomics, continues to produce findings that challenge the idea that cancer is a 'genetic disease', as posited by the somatic mutation theory (SMT). In this prevailing paradigm, tumorigenesis is caused by cancer-driving somatic mutations and clonal expansion. However, results from tumor sequencing, motivated by the genetic paradigm itself, create apparent 'paradoxes' that are not conducive to a pure SMT. But beyond genetic causation, the new results lend credence to old ideas from organismal biology. To resolve inconsistencies between the genetic paradigm of cancer and biological reality, we must complement deep sequencing with deep thinking: embrace formal theory and historicity of biological entities, and (re)consider non-genetic plasticity of cells and tissues. In this Essay, we discuss the concepts of cell state dynamics and tissue fields that emerge from the collective action of genes and of cells in their morphogenetic context, respectively, and how they help explain inconsistencies in the data in the context of SMT.
Collapse
Affiliation(s)
- Sui Huang
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Ana M Soto
- Tufts University School of Medicine, Immunology, Boston, Massachusetts, United States of America
| | - Carlos Sonnenschein
- Tufts University School of Medicine, Immunology, Boston, Massachusetts, United States of America
| |
Collapse
|
40
|
Yamazaki M, Ishimoto T. Targeting Cancer-Associated Fibroblasts: Eliminate or Reprogram? Cancer Sci 2025; 116:613-621. [PMID: 39745128 PMCID: PMC11875776 DOI: 10.1111/cas.16443] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/08/2024] [Accepted: 12/20/2024] [Indexed: 03/05/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are key components of the tumor microenvironment (TME). Given their various roles in tumor progression and treatment resistance, CAFs are promising therapeutic targets in cancer. The elimination of tumor-promoting CAFs has been investigated in various animal models to determine whether it effectively suppresses tumor growth. Based on recent evidence, several simple strategies have been proposed to eliminate tumor-promoting CAFs and attenuate these features. In addition, attention has focused on the critical role that CAFs play in the immunosuppressive TME. Therefore, the functional reprogramming of CAFs in combination with immune checkpoint inhibitors has also been investigated as a possible therapeutic approach. However, although potential targets in CAFs have been widely characterized, the plasticity and heterogeneity of CAFs complicate the understanding of their properties and present difficulties for clinical application. Moreover, the identification of tumor-suppressive CAFs highlights the necessity for the development of therapeutic approaches that can distinguish and switch between tumor-promoting and tumor-suppressive CAFs in an appropriate manner. In this review, we introduce the origins and diversity of CAFs, their role in cancer, and current therapeutic strategies aimed at targeting CAFs, including ongoing clinical evaluations.
Collapse
Affiliation(s)
- Masaya Yamazaki
- Division of CarcinogenesisThe Cancer Institute, Japanese Foundation for Cancer ResearchTokyoJapan
| | - Takatsugu Ishimoto
- Division of CarcinogenesisThe Cancer Institute, Japanese Foundation for Cancer ResearchTokyoJapan
- International Research Center of Medical Sciences (IRCMS)Kumamoto UniversityKumamotoJapan
| |
Collapse
|
41
|
Wu NC, Quevedo R, Nurse M, Hezaveh K, Liu H, Sun F, Muffat J, Sun Y, Simmons CA, McGaha TL, Prinos P, Arrowsmith CH, Ailles L, D'Arcangelo E, McGuigan AP. The use of a multi-metric readout screen to identify EHMT2/G9a-inhibition as a modulator of cancer-associated fibroblast activation state. Biomaterials 2025; 314:122879. [PMID: 39395244 DOI: 10.1016/j.biomaterials.2024.122879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 09/20/2024] [Accepted: 10/04/2024] [Indexed: 10/14/2024]
Abstract
Cancer-associated fibroblasts (CAFs) play a pivotal role in cancer progression, including mediating tumour cell invasion via their pro-invasive secretory profile and ability to remodel the extracellular matrix (ECM). Given that reduced CAF abundance in tumours correlates with improved outcomes in various cancers, we set out to identify epigenetic targets involved in CAF activation in regions of tumour-stromal mixing with the goal of reducing tumour aggressiveness. Using the GLAnCE (Gels for Live Analysis of Compartmentalized Environments) platform, we performed an image-based, phenotypic screen that enabled us to identify modulators of CAF abundance and the capacity of CAFs to induce tumour cell invasion. We identified EHMT2 (also known as G9a), an enzyme that targets the methylation of histone 3 lysine 9 (H3K9), as a potent modulator of CAF abundance and CAF-mediated tumour cell invasion. Transcriptomic and functional analysis of EHMT2-inhibited CAFs revealed EHMT2 participated in driving CAFs towards a pro-invasive phenotype and mediated CAF hyperproliferation, a feature typically associated with activated fibroblasts in tumours. Our study suggests that EHMT2 regulates CAF state within the tumour microenvironment by impacting CAF activation, as well as by magnifying the pro-invasive effects of these activated CAFs on tumour cell invasion through promoting CAF hyperproliferation.
Collapse
Affiliation(s)
- Nila C Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Rene Quevedo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Michelle Nurse
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Kebria Hezaveh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Haijiao Liu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada; Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Fumao Sun
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; The Hospital for Sick Children, Toronto, ON, Canada
| | - Julien Muffat
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; The Hospital for Sick Children, Toronto, ON, Canada
| | - Yu Sun
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada; Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Tracy L McGaha
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Panagiotis Prinos
- Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada
| | - Cheryl H Arrowsmith
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Laurie Ailles
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Elisa D'Arcangelo
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
| | - Alison P McGuigan
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
42
|
Kwon JY, Vera RE, Fernandez-Zapico ME. The multi-faceted roles of cancer-associated fibroblasts in pancreatic cancer. Cell Signal 2025; 127:111584. [PMID: 39756502 PMCID: PMC11807759 DOI: 10.1016/j.cellsig.2024.111584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/13/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025]
Abstract
The tumor microenvironment (TME) has been linked with the pathogenesis of pancreatic ductal adenocarcinoma (PDAC), the most common histological subtype of pancreatic cancer. A central component of the TME are cancer-associated fibroblasts (CAFs), which can either suppress or promote tumor growth in a context-dependent manner. In this review, we will discuss the multi-faceted roles of CAFs in tumor-stroma interactions influencing cancer initiation, progression and therapeutic response.
Collapse
Affiliation(s)
- John Y Kwon
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN 55901, USA.
| | - Renzo E Vera
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN 55901, USA.
| | | |
Collapse
|
43
|
Hernández-Hatibi S, Borau C, Martínez-Bosch N, Navarro P, García-Aznar JM, Guerrero PE. Quantitative characterization of the 3D self-organization of PDAC tumor spheroids reveals cell type and matrix dependence through advanced microscopy analysis. APL Bioeng 2025; 9:016116. [PMID: 40161492 PMCID: PMC11952832 DOI: 10.1063/5.0242490] [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: 10/04/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by an abundant tumor-associated stroma composed from pancreatic stellate cells, which play a critical role in tumor progression. Developing accurate in vitro models requires understanding the complex interactions between tumor cells and their microenvironment. In this study, we present a quantitative imaging-based characterization of the three dimensional (3D) self-organization of PDAC tumour spheroids using a microfluidic platform that mimics key aspects of the tumor microenvironment. Our model incorporates collagen type I hydrogels to recreate the extracellular matrix, activated human pancreatic stellate cells (HPSCs), and various tumor cell types. Advanced imaging techniques, including Lattice Lightsheet Microscopy, allowed us to analyze the 3D growth and spatial organization of the spheroids, revealing intricate biomechanical interactions. Our results indicate that alterations in matrix properties-such as stiffness, pore size, and hydraulic permeability-due to variations in collagen concentration significantly influence the growth patterns and organization of PDAC spheroids, depending on tumor subtype and epithelial-mesenchymal phenotype. Higher collagen concentrations promoted larger spheroids in epithelial-like cell lines, while mesenchymal-type cells required increased collagen for self-organization into smaller spheroids. Furthermore, coculture with HPSCs affected spheroid formation distinctly based on each PDAC cell line's genetic and phenotypic traits. HPSCs had opposing effects on epithelial-like cell lines: one cell line exhibited enhanced spheroid growth, while another showed inhibited formation, whereas mesenchymal-like spheroids showed minimal impact. These results provide insights into tumor-stroma interactions, emphasizing the importance of the cell-specific and matrix-dependent factors for advancing our understanding of PDAC progression and informing future therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Neus Martínez-Bosch
- Cancer Research Program, Hospital del Mar Research Institute (HMRI), Unidad Asociada IIBB-CSIC, 08003 Barcelona, Spain
| | | | | | | |
Collapse
|
44
|
Ebrahim NAA, Soliman SMA. Advanced Biomaterials and Biomedical Devices for Studying Tumor-Associated Fibroblasts: Current Trends, Innovations, and Future Prospects. BIOMEDICAL MATERIALS & DEVICES 2025. [DOI: 10.1007/s44174-025-00287-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/03/2025] [Indexed: 04/23/2025]
|
45
|
Liu T, Wang Q, Geng W, Jiang X, Lu C, Zhang Z, Feng X. Thermosensitive Hydrogel Loaded with α-Mangostin for Enhanced Antitumor Effect of Doxorubicin. ACS Biomater Sci Eng 2025; 11:1000-1012. [PMID: 39884961 DOI: 10.1021/acsbiomaterials.4c02408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
The cancer-associated fibroblasts (CAFs) in tumor stroma present substantial barriers to drug penetration, resulting in tumor resistance and progression. One promising strategy is to reprogram CAFs into a quiescent state, which necessitates novel approaches. Our study introduces a sequential treatment strategy using chitosan thermosensitive hydrogels loaded with α-Mangostin (α-M), a small molecule drug with antifibrotic properties, aimed at reprogramming CAFs within the breast cancer tumor microenvironment (TME). We developed glutathione (GSH)-responsive nanoparticles (NPc) that carry the chemotherapeutic drug doxorubicin (DOX). Treatment with α-M results in the downregulation of CAF-specific biomarkers and a remodeled TME, which improves the penetration of NPc/DOX deep into the tumor tissue. This strategy holds great promise in enhancing cancer therapeutic outcomes in tumors rich in CAFs, particularly in the case of breast cancer.
Collapse
Affiliation(s)
- Tianhui Liu
- Changchun University of Science and Technology, Changchun 130022, Jilin, China
| | - Qingshuang Wang
- Changchun University of Science and Technology, Changchun 130022, Jilin, China
| | - Wenxin Geng
- Changchun University of Science and Technology, Changchun 130022, Jilin, China
| | - Xue Jiang
- Changchun University of Science and Technology, Changchun 130022, Jilin, China
| | - Changshun Lu
- Changchun University of Science and Technology, Changchun 130022, Jilin, China
| | - Zhe Zhang
- Panzhihua University, Panzhihua 617000, Sichuan, China
| | - Xiangru Feng
- Changchun University of Science and Technology, Changchun 130022, Jilin, China
| |
Collapse
|
46
|
Agrawal A, Javanmardi Y, Watson SA, Serwinski B, Djordjevic B, Li W, Aref AR, Jenkins RW, Moeendarbary E. Mechanical signatures in cancer metastasis. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:3. [PMID: 39917412 PMCID: PMC11794153 DOI: 10.1038/s44341-024-00007-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/20/2024] [Indexed: 02/09/2025]
Abstract
The cancer metastatic cascade includes a series of mechanical barrier-crossing events, involving the physical movement of cancer cells from their primary location to a distant organ. This review describes the physical changes that influence tumour proliferation, progression, and metastasis. We identify potential mechanical signatures at every step of the metastatic cascade and discuss some latest mechanobiology-based therapeutic interventions to highlight the importance of interdisciplinary approaches in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Ayushi Agrawal
- Department of Mechanical Engineering, University College London, London, UK
| | - Yousef Javanmardi
- Department of Mechanical Engineering, University College London, London, UK
| | - Sara A. Watson
- Department of Mechanical Engineering, University College London, London, UK
- Division of Biosciences, University College London, London, UK
| | - Bianca Serwinski
- Department of Mechanical Engineering, University College London, London, UK
- Northeastern University London, London, UK
| | - Boris Djordjevic
- Department of Mechanical Engineering, University College London, London, UK
| | - Wenbin Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Amir R. Aref
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Russell W. Jenkins
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
- Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, London, UK
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| |
Collapse
|
47
|
Li B, Shi M, Wang Y, Li P, Yin X, Zhang G, Kang X, Wang H, Gao S, Zheng K, Shi X, Xu X, Zhou Y, Jiang H, Jing W, Guo S, Jin G. A practical distribution pattern of α-SMA-positive carcinoma associated fibroblasts indicates poor prognosis of patients with pancreatic ductal adenocarcinoma. Transl Oncol 2025; 52:102282. [PMID: 39808844 PMCID: PMC11782853 DOI: 10.1016/j.tranon.2025.102282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/08/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
Purpose The present study aimed to clarify the distribution pattern of carcinoma associated fibroblasts (CAFs) across pancreatic ductal adenocarcinoma (PDAC) and its prognostic prediction value. Methods Data of two cohorts were retrospectively collected from consecutive patients who underwent primary pancreatic resection from January 2015 to December 2017. We used tumor specimens to screen out the most suitable markers for the spatial distribution analysis for CAFs subpopulations. We utilized a tissue microarray to assess the spatial intensity of α-SMA expression within the tumor microenvironment. Specifically, we classified CAFs into two types based on their α-SMA spatial expression. Type II CAFs were designated as those located in the juxtatumoural stroma with α-SMA expression that was moderate or higher, and those in the peripheral stroma with α-SMA expression that was less than moderate. All other cases, where the α-SMA expression did not meet these criteria, were categorized as Type I CAFs. Multivariable Cox proportional hazards regression was used to assess risk factors associated with patient outcomes. RNA sequencing data were obtained from bulk tumor samples and isolated CAFs from patients to reveal the distinct pattern and elucidated their fundamental characteristics. Results The α-SMA spatial intensity was the most suitable variable for representative of CAFs spatial characteristics. Patients with Type Ⅰ CAFs were more likely to be allocated into N1 or N2 of the N stage and Ⅱ and Ⅲ of the TNM stage. The spatial distribution pattern of CAFs (Type Ⅰ v.s. Type Ⅱ: HR, 1.568; 95 % CI, 1.053-2.334; P = 0.027) was an independent prognostic factor in the discovery cohort, so as in the validation (Type Ⅰ vs. Type Ⅱ: HR, 2.197; 95 % CI, 1.410-3.422; P = 0.001). RNA sequencing analysis revealed that the differentially expressed genes (DEGs) in Type I CAFs are closely associated with those in corresponding tumor tissues, highlighting the enhanced biological significance of immune-related and oncogenic invasive pathways. Conclusions Our findings that two types of α-SMA-positive CAFs with different spatial patterns present heterogeneously across tissues of PDACs and correlated with patients' outcomes. The spatial location of CAFs may facilitate patients' selection in precision medicine of PDACs.
Collapse
Affiliation(s)
- Bo Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China; Department of Hepatobiliary Pancreatic Surgery, Naval Medical Center, Naval Medical University (Second Military Medical University), 338 West Huaihai Road, Shanghai, 200052, China
| | - Meilong Shi
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Yang Wang
- Department of Pathology, Shanghai Fourth People's Hospital, Tongji University School of Medicine, 1279 Sanmen Road, Shanghai 200434, China
| | - Penghao Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Xiaoyi Yin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Guoxiao Zhang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Xiaochao Kang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Huan Wang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Suizhi Gao
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Kailian Zheng
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Xiaohan Shi
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Xiongfei Xu
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Yukun Zhou
- Department of Hepatobiliary Pancreatic Surgery, Naval Medical Center, Naval Medical University (Second Military Medical University), 338 West Huaihai Road, Shanghai, 200052, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China
| | - Wei Jing
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China.
| | - Shiwei Guo
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China.
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai 200433, China.
| |
Collapse
|
48
|
Ems M, Brichkina A, Lauth M. A safe haven for cancer cells: tumor plus stroma control by DYRK1B. Oncogene 2025; 44:341-347. [PMID: 39863750 PMCID: PMC11790486 DOI: 10.1038/s41388-025-03275-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
The development of resistance remains one of the biggest challenges in clinical cancer patient care and it comprises all treatment modalities from chemotherapy to targeted or immune therapy. In solid malignancies, drug resistance is the result of adaptive processes occurring in cancer cells or the surrounding tumor microenvironment (TME). Future therapy attempts will therefore benefit from targeting both, tumor and stroma compartments and drug targets which affect both sides will be highly appreciated. In this review, we describe a seemingly paradoxical oncogenic mediator with this potential: The dual-specificity tyrosine-phosphorylation regulated kinase 1B (DYRK1B). DYRK1B promotes proliferative quiescence and yet is overexpressed or amplified in many hyperproliferative malignancies including ovarian cancer and pancreatic cancer. In particular the latter disease is a paradigmatic example for a therapy-recalcitrant and highly stroma-rich cancer entity. Here, recent evidence suggests that DYRK1B exerts its oncogenic features by installing a protective niche for cancer cells by directly affecting cancer cells but also the TME. Specifically, DYRK1B not only fosters cell-intrinsic processes like cell survival, chemoresistance, and disease recurrence, but it also upregulates TME and cancer cell-protective innate immune checkpoints and down-modulates anti-tumoral macrophage functionality. In this article, we outline the well-established cell-autonomous roles of DYRK1B and extend its importance to the TME and the control of the tumor immune stroma. In summary, DYRK1B appears as a single novel key player creating a safe haven for cancer cells by acting cell-intrinsically and-extrinsically, leading to the promotion of cancer cell survival, chemoresistance, and relapse. Thus, DYRK1B appears as an attractive drug target for future therapeutic approaches.
Collapse
Affiliation(s)
- Miriam Ems
- Department of Gastroenterology, Endocrinology and Metabolism, Center for Tumor and Immune Biology, Philipps University Marburg, Marburg, Germany
| | - Anna Brichkina
- Institute of Systems Immunology, Philipps University Marburg, Marburg, Germany
| | - Matthias Lauth
- Department of Gastroenterology, Endocrinology and Metabolism, Center for Tumor and Immune Biology, Philipps University Marburg, Marburg, Germany.
| |
Collapse
|
49
|
Du J, Chen X, Xu X, Que Z, Zhai M, Xiang Q, Zhang Z, Zhang Z, Shao Y, Yang X, Miao F, Zhang J, Xie J, Ju S. Enhancing the tissue penetration to improve sonodynamic immunotherapy for pancreatic ductal adenocarcinoma using membrane-camouflaged nanoplatform. Eur J Nucl Med Mol Imaging 2025; 52:1119-1136. [PMID: 39422735 DOI: 10.1007/s00259-024-06952-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
PURPOSE Sonodynamic therapy (SDT) is a promising strategy as an "in situ vaccine" to enhance activation of antitumor immune responses in solid tumors. However, the dense extracellular matrix (ECM) in pancreatic ductal adenocarcinoma (PDAC) lead to hypoxia and limited penetration of most drugs, aggravating the immunosuppressive tumor microenvironment and limiting the efficacy of synergistic sonodynamic immunotherapy. Therefore, it is essential to regulate ECM in order to alleviate tumor hypoxia and enhance the efficacy of sonodynamic immunotherapy for PDAC. METHODS The CPIM nanoplatform, consisting of a macrophage membrane-coated oxygen and drug delivery system (CM@PFOB-ICG-α-Mangostin), was synthesized using ultrasound and extrusion methods. The in vivo homologous targeting and hypoxia alleviation capabilities of CPIM were evaluated through near-infrared (NIR) imaging and photoacoustic (PA) imaging. The tumor growth inhibition potential and ability to reprogram the tumor microenvironment by the CPIM nanoplatform were also investigated. RESULTS Co-delivery of α-Mangostin inhibits CAFs and enhances stromal depletion, thereby facilitating better infiltration of macromolecules. Additionally, the nanoemulsion containing perfluorocarbon (PFC) can target tumor cells and accumulate within them through homologous targeting. The US irradiation results in the rapid release of oxygen, serving as a potential source of sonodynamic therapy for hypoxic tumors. Moreover, CPIM reshapes the immunosuppressive microenvironment increasing the population of cytotoxic T lymphocytes (CTLs), and enhancing their anti-tumor immune response through the use of anti-PDL1 antibodies to block immune checkpoints. CONCLUSION The present study offers a potential strategy for the co-delivery of oxygen and α-Mangostin, aiming to enhance the penetration of tumors to improve SDT. This approach effectively addresses the existing limitations of immune checkpoint blockade (ICB) treatment in solid tumors, while simultaneously boosting the immune response through synergistic sonodynamic immunotherapy.
Collapse
Affiliation(s)
- Jiawei Du
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
| | - Xin Chen
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Medical School, Zhongda Hospital, Southeast University, Nanjing, 210009, P.R. China
| | - Xiaoxuan Xu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
| | - Ziting Que
- Department of Microbiology and Immunology, Medical School, Southeast University, 87th DingJiaQiao Road, Nanjing, 210009, P.R. China
| | - Mengyan Zhai
- Department of Microbiology and Immunology, Medical School, Southeast University, 87th DingJiaQiao Road, Nanjing, 210009, P.R. China
| | - Qinyanqiu Xiang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
| | - Zhiwei Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
| | - Zhiqi Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
| | - Yong Shao
- Department of Microbiology and Immunology, Medical School, Southeast University, 87th DingJiaQiao Road, Nanjing, 210009, P.R. China
| | - Xue Yang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
| | - Fengqin Miao
- Department of Microbiology and Immunology, Medical School, Southeast University, 87th DingJiaQiao Road, Nanjing, 210009, P.R. China
| | - Jianqiong Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
- Department of Microbiology and Immunology, Medical School, Southeast University, 87th DingJiaQiao Road, Nanjing, 210009, P.R. China
| | - Jinbing Xie
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China.
| | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China.
| |
Collapse
|
50
|
Schleinhege R, Neumann I, Oeckinghaus A, Schwab A, Pethő Z. A CNA-35-based high-throughput fibrosis assay reveals ORAI1 as a regulator of collagen release from pancreatic stellate cells. Matrix Biol 2025; 135:70-86. [PMID: 39662708 DOI: 10.1016/j.matbio.2024.12.004] [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: 07/03/2024] [Revised: 11/22/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
RATIONALE Pancreatic stellate cells (PSCs) produce a collagen-rich connective tissue in chronic pancreatitis and pancreatic ductal adenocarcinoma (PDAC). Ca2+-permeable ion channels such as ORAI1 are known to affect PSC proliferation and myofibroblastic phenotype. However, it is unknown whether these channels play a role in collagen secretion. METHODS Using the PSC cell line PS-1, we characterized their cell-derived matrices using staining, mass spectroscopy, and cell migration assays. We developed and validated a high-throughput in vitro fibrosis assay to rapidly determine collagen quantity either with Sirius Red or, in the optimized version, with the collagen-binding peptide CNA-35-tdTomato. We assessed collagen deposition upon stimulating cells with transforming growth factor β1 (TGF-β1) and/or vitamin C without or with ORAI1 modulation. Orai1 expression was assessed by immunohistochemistry in the fibrotic tumor tissue of a murine PDAC model (KPfC). RESULTS We found that TGF-β1 and vitamin C promote collagen deposition from PSCs. We used small interfering RNA (siRNA) and the inhibitor Synta-66 to demonstrate that ORAI1 regulates collagen secretion of PSCs but not NIH-3T3 fibroblasts. Physiological levels of vitamin C induce a drastic increase of the intracellular [Ca2+] in PSCs, with Synta-66 inhibiting Ca2+ influx. Lastly, we revealed Orai1 expression in cancer-associated fibroblasts (CAFs) in murine PDAC (KPfC) samples. CONCLUSION In conclusion, our study introduces a robust in vitro assay for fibrosis and identifies ORAI1 as being engaged in PSC-driven fibrosis.
Collapse
Affiliation(s)
- Rieke Schleinhege
- Institute of Physiology II, University of Münster, Robert-Koch Str. 27B, 48149, Germany
| | - Ilka Neumann
- Institute of Physiology II, University of Münster, Robert-Koch Str. 27B, 48149, Germany
| | - Andrea Oeckinghaus
- Institute of Molecular Tumor Biology, University of Münster, 48149, Germany
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, Robert-Koch Str. 27B, 48149, Germany
| | - Zoltán Pethő
- Institute of Physiology II, University of Münster, Robert-Koch Str. 27B, 48149, Germany.
| |
Collapse
|