1
|
Karim M, Hasan MM, Kim SH, Azam Z, Wahab R, Islam T, Alam F, Kim YJ, Bae DJ, Roy S, Grippo P, Bishehsari F, Choi JU, Al-Hilal TA. Stromal fibrin shapes immune infiltration landscape of pancreatic ductal adenocarcinoma. Biomaterials 2025; 320:123280. [PMID: 40147113 DOI: 10.1016/j.biomaterials.2025.123280] [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: 10/28/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
In pancreatic ductal adenocarcinoma (PDAC), in-situ coagulation creates a thrombotic, crosslinked fibrin (x-fibrin)-rich tumor stroma (FibTS), whose impact on immune cell behavior remains unclear. We aimed to elucidate how FibTS in PDAC regulates immune cell infiltration, polarization, and crosstalk that favors immunosuppressive microenvironment and tumor growth. We assessed the spatial distribution of immune cells by multiplex immunostaining of human PDAC tissues, along with novel bioengineering and mouse tumor models. We investigated how FibTS influences the infiltration of tumor-associated macrophage (TAM) and T-cell subtypes and identified two distinct variants of PDAC, fibrin-high (Fibhi) and fibrin-low (Fiblow). Our findings reveal that PDAC cells secrete fibrinogen and thrombin to form FibTS, which acts as a physical barrier and biochemical niche that restricts CD8+ T-cell and TAM penetration into the tumor. The FibTS impeded immune cell penetration from the tumor stroma into the tumor parenchyma. Selective inhibition of FibTS formation by genetic and pharmacological tools altered the infiltration patterns of CD8+ T-cells and TAMs, decelerating PDAC growth. This study demonstrates that the barrier function of FibTS is crucial for immune evasion, particularly against macrophage and T-cell activity, presenting a potential therapeutic strategy to reshape the immune landscape within PDAC and slow tumor progression.
Collapse
Affiliation(s)
- Mazharul Karim
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Md Mahedi Hasan
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Seung Hyun Kim
- College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea, 02453
| | - Zulfikar Azam
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA
| | - Riajul Wahab
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA
| | - Tamanna Islam
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Farzana Alam
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Yun-Jae Kim
- PrismCDX, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Dong-Jun Bae
- PrismCDX, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Sourav Roy
- Department of Biological Sciences, College of Sciences, University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Paul Grippo
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Faraz Bishehsari
- Gastroenterology Research Center, Division of Gastroenterology, Hepatology & Nutrition, Department of Internal Medicine, University of Texas Houston, TX 77030, USA; MD Anderson Cancer Center-UTHealth Houston Graduate School of Biomedical Sciences, USA
| | - Jeong Uk Choi
- College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea, 02453.
| | - Taslim A Al-Hilal
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
2
|
Fang Y, Tan C, Zheng Z, Yang J, Tang J, Guo R, Silli EK, Chen Z, Chen J, Ge R, Liu Y, Wen X, Liang J, Zhu Y, Jin Y, Li Q, Wang Y. The function of microRNA related to cancer-associated fibroblasts in pancreatic ductal adenocarcinoma. Biochem Pharmacol 2025; 236:116849. [PMID: 40056941 DOI: 10.1016/j.bcp.2025.116849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignant tumor characterized by a poor prognosis. A prominent feature of PDAC is the rich and dense stroma present in the tumor microenvironment (TME), which significantly hinders drug penetration. Cancer-associated fibroblasts (CAFs), activated fibroblasts originating from various cell sources, including pancreatic stellate cells (PSCs) and mesenchymal stem cells (MSCs), play a critical role in PDAC progression and TME formation. MicroRNAs (miRNAs) are small, single-stranded non-coding RNA molecules that are frequently involved in tumorigenesis and progression, exhibiting either oncolytic or oncogenic activity. Increasing evidence suggests that aberrant expression of miRNAs can mediate interactions between cancer cells and CAFs, thereby providing novel therapeutic targets for PDAC treatment. In this review, we will focus on the potential roles of miRNAs that target CAFs or CAFs-derived exosomes in PDAC progression, highlighting the feasibility of therapeutic strategies aimed at restoring aberrantly expressed miRNAs associated with CAFs, offering new pathways for the clinical management of PDAC.
Collapse
Affiliation(s)
- Yaohui Fang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Chunlu Tan
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenjiang Zheng
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianchen Yang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jiali Tang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruizhe Guo
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Epiphane K Silli
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Zhe Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jia Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruyu Ge
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yuquan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xiuqi Wen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jingdan Liang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yunfei Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yutong Jin
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Qian Li
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ying Wang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| |
Collapse
|
3
|
Hunt AL, Randall J, Mansukhani MM, Nyberg K, Nutcharoen A, Davis J, Corgiat B, Mueller C, Melvin S, Sharma M, Johnston L, Swain W, Abulez T, Bateman NW, Maxwell GL, Deeken J, Benyounes A, Petricoin EF, Cannon TL, Conrads TP. Real-time functional proteomics enhances therapeutic targeting in precision oncology molecular tumor boards. NPJ Precis Oncol 2025; 9:111. [PMID: 40234655 PMCID: PMC12000509 DOI: 10.1038/s41698-025-00868-y] [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: 09/20/2024] [Accepted: 03/07/2025] [Indexed: 04/17/2025] Open
Abstract
Collaborative review of molecular profiling data by multidisciplinary molecular tumor boards (MTB) is increasingly important for improving patient management and outcomes, though currently relies nearly exclusively on nucleic acid next-generation sequencing (NGS) and limited panels of immunohistochemistry-based protein abundance data. We examined the feasibility of incorporating real-time laser microdissection (LMD) enrichment of tumor epithelium and commercial CLIA-based reverse phase protein array (RPPA) protein drug target expression/activation profiling into our cancer center's MTB to complement standard clinical NGS-based profiling. The LMD-RPPA workflow was performed within a therapeutically permissive timeframe with a median dwell time of nine days, during which specimens were processed outside of standard clinical workflows. The RPPA-generated data supported additional and/or alternative therapeutic considerations for 54% of profiled patients following review by the MTB. These findings suggest that integrating proteomic/phosphoproteomic and NGS-based genomic data creates opportunities to further personalize clinical decision-making for precision oncology.
Collapse
Affiliation(s)
- Allison L Hunt
- Women's Health Integrated Research Center, Women's Service Line, Inova Health System, 3289 Woodburn Road, Annandale, VA, 22003, USA
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
| | - Jamie Randall
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Drive, Fairfax, VA, 22031, USA
| | - Mahesh M Mansukhani
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Kara Nyberg
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Drive, Fairfax, VA, 22031, USA
| | - Aratara Nutcharoen
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Drive, Fairfax, VA, 22031, USA
- Department of Pathology, Inova Fairfax Hospital, 3300 Gallows Road, Falls Church, VA, 22042, USA
| | - Justin Davis
- Ignite Proteomics Inc., 15000 W 6th Avenue, Golden, CO, 80401, USA
| | - Brian Corgiat
- Ignite Proteomics Inc., 15000 W 6th Avenue, Golden, CO, 80401, USA
| | - Claudius Mueller
- Ignite Proteomics Inc., 15000 W 6th Avenue, Golden, CO, 80401, USA
| | - Savannah Melvin
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Drive, Fairfax, VA, 22031, USA
| | - Meenakshi Sharma
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Drive, Fairfax, VA, 22031, USA
| | - Laura Johnston
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Drive, Fairfax, VA, 22031, USA
| | - Whitney Swain
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Drive, Fairfax, VA, 22031, USA
| | - Tamara Abulez
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Nicholas W Bateman
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - G Larry Maxwell
- Women's Health Integrated Research Center, Women's Service Line, Inova Health System, 3289 Woodburn Road, Annandale, VA, 22003, USA
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
| | - John Deeken
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Drive, Fairfax, VA, 22031, USA
| | - Amin Benyounes
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Drive, Fairfax, VA, 22031, USA
| | - Emanuel F Petricoin
- Ignite Proteomics Inc., 15000 W 6th Avenue, Golden, CO, 80401, USA
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA, 20110, USA
| | - Timothy L Cannon
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Drive, Fairfax, VA, 22031, USA.
| | - Thomas P Conrads
- Women's Health Integrated Research Center, Women's Service Line, Inova Health System, 3289 Woodburn Road, Annandale, VA, 22003, USA.
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA.
| |
Collapse
|
4
|
Han D, Sojic N, Jiang D. Spatial Profiling of Multiple Enzymatic Activities at Single Tissue Sections via Fenton-Promoted Electrochemiluminescence. J Am Chem Soc 2025; 147:9610-9619. [PMID: 40063963 DOI: 10.1021/jacs.4c17749] [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]
Abstract
Profiling multiple enzymatic activities in tissue is crucial for understanding complex metabolic and signaling networks, yet remains a challenge with existing optical microscopies. Here, we developed a Fenton-promoted luminol electrochemiluminescence (ECL) imaging method to achieve the spatial mapping of multiple enzymatic activities within a single tissue section. This method quantitatively visualizes individual enzymatic activity by combining the enzymatic conversion of substrates with the chemical confinement of the locally produced hydrogen peroxide. To achieve high-resolution spatial imaging by limiting the diffusion (∼500 μm) of hydrogen peroxide, iron oxide nanoparticles were coated on the tissue surface to initiate the Fenton process, locally converting hydrogen peroxide into short-lived hydroxyl radicals with a nanometer-scale diffusion range. The Fenton-promoted ECL emission is confined at the enzymatic conversion sites, offering unprecedented spatial visualization of four tumor-associated oxidases within a single tissue section. Colocalization revealed a synergistic effect between lysyl oxidase and quiescin sulfhydryl oxidase on post-translational modifications of tumor extracellular matrix proteins, along with a previously undiscovered interaction with amiloride-sensitive amine oxidase, which could not be distinguished based on expressions or single enzymatic activity alone. This approach offers a novel activity-based protein profiling tool at the tissue level, providing new data for future enzynomic research and multimodal imaging.
Collapse
Affiliation(s)
- Dongni Han
- State Key Laboratory of Analytical Chemistry for Life Science and School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210093, China
| | - Neso Sojic
- University of Bordeaux, CNRS, Bordeaux INP, ISM, UMR, 5255, F-33400 Talence, France
| | - Dechen Jiang
- State Key Laboratory of Analytical Chemistry for Life Science and School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210093, China
| |
Collapse
|
5
|
Wei JR, Lu MY, Wei TH, Fleishman JS, Yu H, Chen XL, Kong XT, Sun SL, Li NG, Yang Y, Ni HW. Overcoming cancer therapy resistance: From drug innovation to therapeutics. Drug Resist Updat 2025; 81:101229. [PMID: 40081221 DOI: 10.1016/j.drup.2025.101229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
One of the major limitations of cancer therapy is the emergence of drug resistance. This review amis to provide a focused analysis of the multifactorial mechanisms underlying therapy resistance,with an emphasis on actionable insights for developing novel therapeutic strategies. It concisely outlines key factors contributing to therapy resistance, including drug delivery barriers, cancer stem cells (CSCs), epithelial-mesenchymal transition (EMT), cancer heterogeneity, tumor microenvironment (TME), genetic mutations, and alterlations in gene expression. Additionally, we explore how tumors evade targeted therapies through pathway-specific mechanisms that restore disrupted signaling pathways. The review critically evaluates innovative strategies designed to sensitize resistant tumor cells, such as targeted protein dedgradation, antibody-drug conjugates, structure-based drug design, allosteric drugs, multitarget drugs, nanomedicine and others We also highlight the importance of understanding the pharmacological actions of these agents and their integration into treatment regimens. By synthesizing current knowledge and identifying gaps in our understanding, this review aims to guide future research and improve patient outcomes in cancer therapy.
Collapse
Affiliation(s)
- Jin-Rui Wei
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China; The First Clinical College of Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Meng-Yi Lu
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Tian-Hua Wei
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Joshua S Fleishman
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Hui Yu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Xiao-Li Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Xiang-Tu Kong
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Shan-Liang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China.
| | - Nian-Guang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Ye Yang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Hai-Wen Ni
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China.
| |
Collapse
|
6
|
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
|
7
|
Qu C, Zeng P, Li C, Hu W, Yang D, Wang H, Yuan H, Cao J, Xiu D. A machine learning model based on preoperative multiparametric quantitative DWI can effectively predict the survival and recurrence risk of pancreatic ductal adenocarcinoma. Insights Imaging 2025; 16:38. [PMID: 39962007 PMCID: PMC11833029 DOI: 10.1186/s13244-025-01915-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/26/2025] [Indexed: 02/20/2025] Open
Abstract
PURPOSE To develop a machine learning (ML) model combining preoperative multiparametric diffusion-weighted imaging (DWI) and clinical features to better predict overall survival (OS) and recurrence-free survival (RFS) following radical surgery for pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS A retrospective analysis was conducted on 234 PDAC patients who underwent radical resection at two centers. Among 101 ML models tested for predicting postoperative OS and RFS, the best-performing model was identified based on comprehensive evaluation metrics, including C-index, Brier scores, AUC curves, clinical decision curves, and calibration curves. This model's risk stratification capability was further validated using Kaplan-Meier survival analysis. RESULTS The random survival forest model achieved the highest C-index (0.828/0.723 for OS and 0.781/0.747 for RFS in training/validation cohorts). Incorporating nine key factors-D value, T-stage, ADC-value, postoperative 7th day CA19-9 level, AJCC stage, tumor differentiation, type of operation, tumor location, and age-optimized the model's predictive accuracy. The model had integrated Brier score below 0.13 and C/D AUC values above 0.85 for both OS and RFS predictions. It also outperformed traditional models in predictive ability and clinical benefit, as shown by clinical decision curves. Calibration curves confirmed good predictive consistency. Using cut-off scores of 16.73/29.05 for OS/RFS, Kaplan-Meier analysis revealed significant prognostic differences between risk groups (p < 0.0001), highlighting the model's robust risk prediction and stratification capabilities. CONCLUSION The random survival forest model, combining DWI and clinical features, accurately predicts survival and recurrence risk after radical resection of PDAC and effectively stratifies risk to guide clinical treatment. CRITICAL RELEVANCE STATEMENT The construction of 101 ML models based on multiparametric quantitative DWI combined with clinical variables has enhanced the prediction performance for survival and recurrence risks in patients undergoing radical resection for PDAC. KEY POINTS This study first develops DWI-based radiological-clinical ML models predicting PDAC prognosis. Among 101 models, RFS is the best and outperforms other traditional models. Multiparametric DWI is the key prognostic predictor, with model interpretations through SurvSHAP.
Collapse
Affiliation(s)
- Chao Qu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Piaoe Zeng
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Changlei Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Weiyu Hu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dongxia Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hangyan Wang
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Huishu Yuan
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Jingyu Cao
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Dianrong Xiu
- Department of General Surgery, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
8
|
Chen X, Miao J, Huang X, Han X, Zheng L, Chen J, Chen L, Li L. Multiphoton imaging-based quantifiable collagen signatures for predicting outcomes in patients with pancreatic ductal adenocarcinoma. Sci Rep 2025; 15:4414. [PMID: 39910233 PMCID: PMC11799447 DOI: 10.1038/s41598-025-88984-4] [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: 11/01/2024] [Accepted: 02/03/2025] [Indexed: 02/07/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents a clinical challenge due to its poor prognosis and high mortality rate. Here, we aimed to enhance the prognostic prediction of patients with PDAC by studying collagen features in tumor microenvironment using multiphoton microscopy (MPM) combining with image processing technique. We identified eight distinct tumor-associated collagen signatures (TACS1-8) from multiphoton images of PDAC tissues and developed an optical biomarker, TACS-score, based on the TACS1-8 using ridge regression analysis. Additionally, we also extracted 142 microscopic TACS (M-TACS) from second-harmonic generation (SHG) images and constructed a new robust biomarker, M-TACS-score, using the least absolute shrinkage and selection operator (LASSO) regression analysis. Our statistical results demonstrate that as two new optical biomarkers, TACS- and M-TACS-score, are independent prognostic factors and have good discriminatory ability (high AUC) as well as risk stratification (high HR) comparing with traditional clinical model (combining seven clinical risk factors, age, sex, TNM stage, tumor location and differentiation, perineural and lymph-vascular invasion) in predicting overall survival (OS) of patients with PDAC, highlighting their potential prognostic and predictive value. A combination of label-free multiphoton imaging technique and computer-aided image processing method may offer a novel and promising approach for finding new biomarkers to improve prognosis prediction and thereby tailor treatment strategies more effectively.
Collapse
Affiliation(s)
- Xiwen Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Jikui Miao
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Xingxin Huang
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Xiahui Han
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Liqin Zheng
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Jianxin Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China
| | - Linying Chen
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, China.
| | - Lianhuang Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, 350007, China.
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
Porter G, Norris MD, Apte M, Merlot AM. Spatial profiling of endoplasmic reticulum stress markers in tumor associated cells predicts patient outcomes in pancreatic cancer. Neoplasia 2025; 60:101115. [PMID: 39818177 PMCID: PMC11786694 DOI: 10.1016/j.neo.2024.101115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
INTRODUCTION The impact of endoplasmic reticulum (ER) stress in tumor-associated cells, such as cancer associated fibroblasts (CAFs), immune cells and endothelial cells, on patient outcomes in clinical specimens have not been examined. For the first time, we characterized the expression and spatial locations of ER stress markers, BiP and CHOP, in tumor-associated cells and assessed their prognostic significance in a panel of pancreatic ductal adenocarcinoma (PDAC) patient samples. METHODS Multiplex immunofluorescence was performed on tumor microarrays and images were analyzed using HALO AI software. RESULTS BiP and CHOP were upregulated in CAFs and endothelial cells in PDAC sections relative to non-neoplastic pancreas sections. High BiP expression in CAFs and endothelial cells was associated with greater vascular invasion and in immune cells was correlated with increased tumor size. High CHOP expression in immune cells correlated with poor patient survival. CAFs and immune cells were more likely to express BiP or CHOP when located close (< 20 μm) to tumor cells. High expression of CHOP in CAFs close to tumor cells correlated with improved patient survival. CONCLUSION For the first time, this study demonstrated that ER stress occurs in CAFs and immune cells predominantly in proximity to tumor cells in PDAC patient tissue. The correlation of high ER stress in immune cells with poor patient survival highlights the importance of the TME and the use of spatial analysis for the identification of novel biomarkers.
Collapse
Affiliation(s)
- Georgia Porter
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia; School of Clinical Medicine, Faculty of Medicine & Health, University of New South Wales, Kensington, New South Wales 2031, Australia; UNSW Centre for Childhood Cancer Research, Faculty of Medicine &Health, University of New South Wales, Kensington, New South Wales 2031, Australia
| | - Murray D Norris
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia; School of Clinical Medicine, Faculty of Medicine & Health, University of New South Wales, Kensington, New South Wales 2031, Australia; UNSW Centre for Childhood Cancer Research, Faculty of Medicine &Health, University of New South Wales, Kensington, New South Wales 2031, Australia
| | - Minoti Apte
- Pancreatic Research Group, South Western Sydney Clinical Campuses, Faculty of Medicine and Health, UNSW Sydney, NSW 2052, Australia; Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia
| | - Angelica M Merlot
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia; School of Clinical Medicine, Faculty of Medicine & Health, University of New South Wales, Kensington, New South Wales 2031, Australia; UNSW Centre for Childhood Cancer Research, Faculty of Medicine &Health, University of New South Wales, Kensington, New South Wales 2031, Australia; Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2031, Australia.
| |
Collapse
|
13
|
Peng J, Sun J, Yu Y, Yuan Q, Zhang Y. Integrative multi-omics analysis reveals the role of toll-like receptor signaling in pancreatic cancer. Sci Rep 2025; 15:52. [PMID: 39747201 PMCID: PMC11696379 DOI: 10.1038/s41598-024-84062-3] [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: 05/04/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
As one of the most destructive and invasive cancers, pancreatic cancer exhibits complex tumor heterogeneity, which has been a major challenge for clinicians in terms of patient treatment and prognosis. The toll-like receptor (TLR) pathway is closely related to the immune microenvironment within various cancer tissues. To explore the development pattern of pancreatic cancer and find an ideal biomarker, our research has explored the mechanism of the TLR pathway in pancreatic cancer. We collected single-cell expression data from 57,024 cells and transcriptomic data from 945 pancreatic cancer patients, and conducted a series of analyses at both the single-cell and transcriptomic levels. By calculating the TLR pathway score, we clustered pancreatic cancer patients and conducted a series of analyses including metabolic pathways, immune microenvironment, drug sensitivity and so on. In the process of building prognostic models, we screened 33 core genes related to the prognosis of pancreatic cancer, and combined a series of machine learning algorithms to build the prognosis model of pancreatic cancer. We used single cell sequencing to clarify the complex intrinsic relationship between TLR pathway and pancreatic cancer. The strongest TLR signals were observed in macrophages and endothelial cells. With the occurrence of pancreatic cancer, the TLR signal of various cell types gradually increased, but with the increase of the malignant degree of ductal epithelial cells, the TLR signal gradually weakened. Cluster analysis showed that patients with the most active TLR pathway had severe dysregulation of immune microenvironment and the worst prognosis. Finally, we combined a series of machine learning algorithms to build a pancreatic cancer prognosis model that includes four genes (NT5E, TGFBI, ANLN, and FAM83A). The model showed strong performance in predicting the survival state of pancreatic cancer samples. We explored the important role of TLR pathway in pancreatic cancer and established and validated a new prognosis model for pancreatic cancer based on TLR-related genes.
Collapse
Affiliation(s)
- Jie Peng
- Ningde Clinical Medical College of Fujian Medical University, Fujian, China
- Ningde Municipal Hospital of Ningde Normal University, Fujian, China
| | - Jiaao Sun
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Youfeng Yu
- Ningde Clinical Medical College of Fujian Medical University, Fujian, China
- Ningde Municipal Hospital of Ningde Normal University, Fujian, China
| | - Qihang Yuan
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Yong Zhang
- Ningde Clinical Medical College of Fujian Medical University, Fujian, China.
- Ningde Municipal Hospital of Ningde Normal University, Fujian, China.
| |
Collapse
|
14
|
Tang R, Liu M, Shu Q, Chen X, Cai L. Performance of fibroblast activating protein inhibitor PET imaging for pancreatic neoplasms assessment: a systematic review and meta-analysis. Eur Radiol 2024; 34:7804-7812. [PMID: 38907099 DOI: 10.1007/s00330-024-10843-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/05/2024] [Accepted: 04/21/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Recent studies have shown the potential of fibroblast activating protein inhibitor (FAPI) PET imaging for pancreatic cancer assessment. PURPOSE This article is dedicated to comparing the diagnostic efficacy of FAPI PET and [18F]fluorodeoxyglucose (FDG) PET in the evaluation of primary tumors, lymph nodes, and distant metastases in pancreatic cancer. METHODS In this review, we conducted a systematic search of studies published in PubMed and Web of Science databases up to September 18, 2023. All included studies used radionuclide labeled FAPI and FDG as PET diagnostic tracers to evaluate their applicability in patients with pancreatic cancer. RESULTS The FAPI PET imaging group showed significantly higher sensitivity in the detection of primary lesions (1.000, [95% CI: 0.999-1.000]), lymph node metastases (0.624 [95% CI: 0.391-0.834]) and distant metastatic (0.965 [95% CI: 0.804-1.000]) in pancreatic cancer compared to the FDG PET imaging group (0.889 [95% CI: 0.788-0.966], 0.373 [95% CI: 0.163-0.606] and 0.889 [95% CI: 0.689-0.999], respectively). Furthermore, the maximum standardized uptake value (SUVmax) in FAPI PET imaging is significantly higher than that in FDG imaging for primary lesions (mean difference (MD) = 7.51, 95% CI: 5.34-9.67). CONCLUSION Compared with [18F]FDG PET/CT, FAPI PET imaging showed higher sensitivity, SUVmax. This method can be effectively utilized for the evaluation of pancreatic cancer. CLINICAL RELEVANCE STATEMENT Fibroblast activating protein inhibitor PET may be a better alternative to [18F]FDG in evaluating primary pancreatic cancer, lymph node metastases, and distant metastases. KEY POINTS Fibroblast activating protein inhibitor (FAPI) PET is compared with FDG PET for evaluating pancreatic cancer. Multiple radiolabeled FAPI variants have shown promising results in the diagnosis of pancreatic cancer. FAPI PET imaging effectively helps clinicians diagnose and stage pancreatic cancer.
Collapse
Affiliation(s)
- Ranbie Tang
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, 646000, Luzhou, Sichuan, PR China
- Institute of Nuclear Medicine, Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
| | - Mengna Liu
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, 646000, Luzhou, Sichuan, PR China
- Institute of Nuclear Medicine, Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
| | - Qiaoqiao Shu
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, 646000, Luzhou, Sichuan, PR China
- Institute of Nuclear Medicine, Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
| | - Xi Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, 646000, Luzhou, Sichuan, PR China
- Institute of Nuclear Medicine, Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China
| | - Liang Cai
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China.
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, 646000, Luzhou, Sichuan, PR China.
- Institute of Nuclear Medicine, Southwest Medical University, No. 25, Taiping St, 646000, Luzhou, Sichuan, PR China.
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, 400010, Chongqing, PR China.
| |
Collapse
|
15
|
Zhang R, Peng J, Zhang Y, Zheng K, Chen Y, Liu L, Li T, Liu J, Li Y, Yang S, Wang M, Cui M, Zhang X, Gao J, Kleeff J, Liao Q, Liu Q. Pancreatic cancer cell-derived migrasomes promote cancer progression by fostering an immunosuppressive tumor microenvironment. Cancer Lett 2024; 605:217289. [PMID: 39389157 DOI: 10.1016/j.canlet.2024.217289] [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: 05/29/2024] [Revised: 10/01/2024] [Accepted: 10/06/2024] [Indexed: 10/12/2024]
Abstract
Pancreatic cancer is distinguished by an immunosuppressive tumor microenvironment (TME) that facilitates cancer progression. The assembly of the TME involves numerous contributing factors. Migrasomes, recently identified as cellular organelles in migrating cells, play a pivotal role in intercellular signaling. However, research into their involvement in cancers remains nascent. Thus far, whether pancreatic cancer cells generate migrasomes and their potential role in TME formation remains unexplored. In this study, it was found that both murine and human pancreatic cancer cells could indeed generate migrasomes, termed pancreatic cancer cell-derived migrasomes (PCDMs), which actively promote cancer progression. Moreover, utilizing chemokine antibody arrays and quantitative mass spectrometry analysis, we observed significant differences between the chemokines, cytokines, and proteins present in PCDMs compared to their originating cell bodies. Notably, PCDMs exhibited an enrichment of immunosuppression-inducing factors. Furthermore, macrophages could directly uptake PCDMs, leading to the expression of high levels of M2-like markers and secretion of tumor-promoting factors. PCDM-induced macrophages played a pivotal role in inhibiting T cell proliferation and activation partially through ARG-1. In summary, this study provides compelling evidence that pancreatic cancer cells generate migrasomes, which play a crucial role in promoting tumor progression by contributing to an immunosuppressive TME. The exploration of migrasomes as a therapeutic target could pave the way for the development of tailored immunotherapies for pancreatic cancer.
Collapse
Affiliation(s)
- Ronghua Zhang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China; Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junya Peng
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Yalu Zhang
- Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Kexin Zheng
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yang Chen
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Lulu Liu
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Tong Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Shanghai Pancreatic Cancer Institute, Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jingkai Liu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Ying Li
- Cryo-EM Facility at Technology Center for Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Sen Yang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengyi Wang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming Cui
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiang Zhang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junyi Gao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jorg Kleeff
- Department of Visceral, Vascular and Endocrine Surgery, Martin-Luther-University Halle-Wittenberg, University Medical Center Halle (Saale), Halle (Saale), Germany
| | - Quan Liao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Qiaofei Liu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
16
|
Pang N, Yang Z, Zhang W, Du Y, Zhang L, Li X, Peng Y, Qi X. Cancer-associated fibroblasts barrier breaking via TGF-β blockade paved way for docetaxel micelles delivery to treat pancreatic cancer. Int J Pharm 2024; 665:124706. [PMID: 39277152 DOI: 10.1016/j.ijpharm.2024.124706] [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: 05/29/2024] [Revised: 08/31/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
TGF-β is a crucial regulator in tumor microenvironment (TME), especially for myofibroblastic cancer-associated fibroblasts (myCAFs). The myCAFs can be motivated by TGF-β signaling to erect pro-tumor TME, meanwhile, myCAFs overexpress TGF-β to mediate the crosstalk between tumor and stromal cells. The blockade of TGF-β can break cancer-associated fibroblasts barrier, consequently opening the access for drugs into tumor. The TGF-β is a promising target in anti-tumor therapy. Herein, we introduced a two-stage combination therapy (TC-Therapy), including TGF-β receptor I inhibitor SB525334 (SB) and cytotoxicity agent docetaxel micelle (DTX-M). We found that SB and DTX-M synergistically inhibited myCAFs proliferation and elevated p53 protein expression in BxPC-3/3T3 mixed cells. Gene and protein tests demonstrated that SB cut off TGF-β signaling via receptor blockade and it did not arouse TGF-β legend compensated internal autocrine. On the contrary, two agents combined decreased TGF-β secretion and inhibited myCAFs viability marked by α-SMA and FAPα. TC-Therapy was applied in BxPc-3/3T3 mixed tumor-bearing mice model. After TC-Therapy, the α-SMA+/ FAPα+ myCAFs faded increasingly and collagenous fibers mainly secreted by myCAFs decreased dramatically as well. More than that, the myCAFs barrier breaking helped to normalize micro-vessels and paved way for micelle penetration. The TGF-β protein level of TC-Therapy in TME was much lower than that of simplex DTX-M, which might account for TME restoration. In conclusion, TGF-β inhibitor acted as the pioneer before nano chemotherapeutic agents. The TC-Therapy of TGF-β signaling inhibition and anti-tumor agent DTX-M is a promising regimen without arising metastasis risk to treat pancreatic cancer. The therapeutic regimen focused on TGF-β related myCAFs reminds clinicians to have a comprehensive understanding of pancreatic cancer.
Collapse
Affiliation(s)
- Ning Pang
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Zhenzhen Yang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wenjie Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yitian Du
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Lu Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xin Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yiwei Peng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xianrong Qi
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
17
|
Lee TS, Lee SH, Kim J, Lee MH, Cho IR, Ryu JK, Kim YT, Paik WH. Increased needle passes for comparable diagnostic yield in endoscopic ultrasound-guided tissue acquisition for pancreatic stiff lesions measured by elastography. Pancreatology 2024; 24:1192-1198. [PMID: 39277479 DOI: 10.1016/j.pan.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/15/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND/OBJECTIVES Pancreatic cancer is characterized by tissue stiffness due to the high concentration of cancer-associated fibroblasts and extracellular matrix. Endoscopic ultrasound-guided tissue acquisition (EUS-TA) is performed to diagnose pancreatic cancer but yields false-negative results attributed to inadequate specimens. EUS-elastography is a real-time assessment method to pancreatic tissue stiffness. This study aims to investigate the correlation between diagnostic yield and the number of needle passes based on the stiffness measured by elastography. METHODS Patients who underwent EUS-TA for pancreatic solid mass were retrospectively reviewed and included in this study. The number of needle passes during EUS-TA was determined based on macroscopic on-site evaluation. Tissue stiffness measurements were taken using EUS-elastography. The primary study outcome was the diagnostic yield. The secondary outcome included the number of needle passes required for a diagnosis. RESULTS A total of 652 patients were included. The average stiffness differed depending on the location of the tumor, and high-stiffness group had more malignant lesions. Although the diagnostic yield was not significantly different between groups, the number of needle passes was significantly higher in the high-stiffness group (3.6 ± 1.0 vs. 3.2 ± 0.9, p < 0.001). CONCLUSIONS The higher the stiffness of the pancreatic mass in EUS-elastography, the more needle passes are required to achieve a comparable diagnostic yield.
Collapse
Affiliation(s)
- Tae Seung Lee
- Department of Internal Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-Gu, Seoul, Republic of Korea
| | - Sang Hyub Lee
- Department of Internal Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-Gu, Seoul, Republic of Korea
| | - Junyeol Kim
- Department of Internal Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-Gu, Seoul, Republic of Korea
| | - Myeong Hwan Lee
- Department of Internal Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-Gu, Seoul, Republic of Korea
| | - In Rae Cho
- Department of Internal Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-Gu, Seoul, Republic of Korea
| | - Ji Kon Ryu
- Department of Internal Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-Gu, Seoul, Republic of Korea
| | - Yong-Tae Kim
- Department of Internal Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-Gu, Seoul, Republic of Korea
| | - Woo Hyun Paik
- Department of Internal Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-Gu, Seoul, Republic of Korea.
| |
Collapse
|
18
|
Xia C, Qu JR, Jiao YP, Lu CQ, Zhao B, Ge RJ, Qiu Y, Cao BY, Yu Q, Xia TY, Meng XP, Song Y, Zhang LH, Long XY, Ye J, Ding ZM, Cai W, Ju SH. Signal enhancement ratio of multi-phase contrast-enhanced MRI: an imaging biomarker for survival in pancreatic adenocarcinoma. Eur Radiol 2024; 34:7460-7470. [PMID: 38750169 PMCID: PMC11519106 DOI: 10.1007/s00330-024-10746-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/07/2024] [Accepted: 03/30/2024] [Indexed: 10/29/2024]
Abstract
OBJECTIVES To evaluate signal enhancement ratio (SER) for tissue characterization and prognosis stratification in pancreatic adenocarcinoma (PDAC), with quantitative histopathological analysis (QHA) as the reference standard. METHODS This retrospective study included 277 PDAC patients who underwent multi-phase contrast-enhanced (CE) MRI and whole-slide imaging (WSI) from three centers (2015-2021). SER is defined as (SIlt - SIpre)/(SIea - SIpre), where SIpre, SIea, and SIlt represent the signal intensity of the tumor in pre-contrast, early-, and late post-contrast images, respectively. Deep-learning algorithms were implemented to quantify the stroma, epithelium, and lumen of PDAC on WSIs. Correlation, regression, and Bland-Altman analyses were utilized to investigate the associations between SER and QHA. The prognostic significance of SER on overall survival (OS) was evaluated using Cox regression analysis and Kaplan-Meier curves. RESULTS The internal dataset comprised 159 patients, which was further divided into training, validation, and internal test datasets (n = 60, 41, and 58, respectively). Sixty-five and 53 patients were included in two external test datasets. Excluding lumen, SER demonstrated significant correlations with stroma (r = 0.29-0.74, all p < 0.001) and epithelium (r = -0.23 to -0.71, all p < 0.001) across a wide post-injection time window (range, 25-300 s). Bland-Altman analysis revealed a small bias between SER and QHA for quantifying stroma/epithelium in individual training, validation (all within ± 2%), and three test datasets (all within ± 4%). Moreover, SER-predicted low stromal proportion was independently associated with worse OS (HR = 1.84 (1.17-2.91), p = 0.009) in training and validation datasets, which remained significant across three combined test datasets (HR = 1.73 (1.25-2.41), p = 0.001). CONCLUSION SER of multi-phase CE-MRI allows for tissue characterization and prognosis stratification in PDAC. CLINICAL RELEVANCE STATEMENT The signal enhancement ratio of multi-phase CE-MRI can serve as a novel imaging biomarker for characterizing tissue composition and holds the potential for improving patient stratification and therapy in PDAC. KEY POINTS Imaging biomarkers are needed to better characterize tumor tissue in pancreatic adenocarcinoma. Signal enhancement ratio (SER)-predicted stromal/epithelial proportion showed good agreement with histopathology measurements across three distinct centers. Signal enhancement ratio (SER)-predicted stromal proportion was demonstrated to be an independent prognostic factor for OS in PDAC.
Collapse
Affiliation(s)
- Cong Xia
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Jin-Rong Qu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Yi-Ping Jiao
- Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, China
| | - Chun-Qiang Lu
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Ben Zhao
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Rong-Jun Ge
- School of Instrument Science and Engineering, Southeast University, Nanjing, China
| | - Yue Qiu
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Bu-Yue Cao
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Qian Yu
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Tian-Yi Xia
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Xiang-Pan Meng
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China
| | - Yang Song
- MR Scientific Marketing, Siemens Healthineers Ltd., Shanghai, China
| | - Li-Hua Zhang
- Department of Pathology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xue-Ying Long
- Department of Radiology, The Xiangya Hospital of Central South University, Changsha, China
| | - Jing Ye
- Department of Medical Imaging, Subei People's Hospital, Medical School of Yangzhou University, Yangzhou, China
| | - Zhi-Min Ding
- Department of Radiology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Wu Cai
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Sheng-Hong Ju
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu, China.
| |
Collapse
|
19
|
Zhao Y, Qin C, Lin C, Li Z, Zhao B, Li T, Zhang X, Wang W. Pancreatic ductal adenocarcinoma cells reshape the immune microenvironment: Molecular mechanisms and therapeutic targets. Biochim Biophys Acta Rev Cancer 2024; 1879:189183. [PMID: 39303859 DOI: 10.1016/j.bbcan.2024.189183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/23/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a digestive system malignancy characterized by challenging early detection, limited treatment alternatives, and generally poor prognosis. Although there have been significant advancements in immunotherapy for hematological malignancies and various solid tumors in recent decades, with impressive outcomes in recent preclinical and clinical trials, the effectiveness of these therapies in treating PDAC continues to be modest. The unique immunological microenvironment of PDAC, especially the abnormal distribution, complex composition, and variable activation states of tumor-infiltrating immune cells, greatly restricts the effectiveness of immunotherapy. Undoubtedly, integrating data from both preclinical models and human studies helps accelerate the identification of reliable molecules and pathways responsive to targeted biological therapies and immunotherapies, thereby continuously optimizing therapeutic combinations. In this review, we delve deeply into how PDAC cells regulate the immune microenvironment through complex signaling networks, affecting the quantity and functional status of immune cells to promote immune escape and tumor progression. Furthermore, we explore the multi-modal immunotherapeutic strategies currently under development, emphasizing the transformation of the immunosuppressive environment into an anti-tumor milieu by targeting specific molecular and cellular pathways, providing insights for the development of novel treatment strategies.
Collapse
Affiliation(s)
- Yutong Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Cheng Qin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Chen Lin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Zeru Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Bangbo Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Tianyu Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Xiangyu Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Weibin Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| |
Collapse
|
20
|
Espona-Fiedler M, Patthey C, Lindblad S, Sarró I, Öhlund D. Overcoming therapy resistance in pancreatic cancer: New insights and future directions. Biochem Pharmacol 2024; 229:116492. [PMID: 39153553 DOI: 10.1016/j.bcp.2024.116492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Pancreatic adenocarcinoma (PDAC) is predicted to become the second leading cause of cancer deaths by 2030 and this is mostly due to therapy failure. Limited treatment options and resistance to standard-of-care (SoC) therapies makes PDAC one of the cancer types with poorest prognosis and survival rates [1,2]. Pancreatic tumors are renowned for their poor response to therapeutic interventions including targeted therapies, chemotherapy and radiotherapy. Herein, we review hallmarks of therapy resistance in PDAC and current strategies aiming to tackle escape mechanisms and to re-sensitize cancer cells to therapy. We will further provide insights on recent advances in the field of drug discovery, nanomedicine, and disease models that are setting the ground for future research.
Collapse
Affiliation(s)
- Margarita Espona-Fiedler
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå Universitet, Umeå, Sweden.
| | - Cedric Patthey
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå Universitet, Umeå, Sweden
| | - Stina Lindblad
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden
| | - Irina Sarró
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Universitat de Barcelona, Barcelona, Spain
| | - Daniel Öhlund
- Department of Diagnostic and Intervention, Umeå Universitet, Umeå, Sweden; Wallenberg Centre for Molecular Medicine, Umeå Universitet, Umeå, Sweden.
| |
Collapse
|
21
|
Li J, Zhang W, Chen L, Wang X, Liu J, Huang Y, Qi H, Chen L, Wang T, Li Q. Targeting extracellular matrix interaction in gastrointestinal cancer: Immune modulation, metabolic reprogramming, and therapeutic strategies. Biochim Biophys Acta Rev Cancer 2024; 1879:189225. [PMID: 39603565 DOI: 10.1016/j.bbcan.2024.189225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/17/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
The extracellular matrix (ECM) is a major constituent of the tumor microenvironment, acting as a mediator that supports the progression of gastrointestinal (GI) cancers, particularly in mesenchymal subtypes. Beyond providing structural support, the ECM actively shapes the tumor microenvironment (TME) through complex biochemical and biomechanical remodeling. Dysregulation of ECM composition and signaling is closely linked to increased cancer aggressiveness, poor prognosis, and resistance to therapy. ECM components, such as collagen, fibronectin, laminin, and periostin, influence tumor growth, metastasis, immune modulation, and metabolic reprogramming by interacting with tumor cells, immune cells, and cancer-associated fibroblasts. In this review, we highlight the heterogeneous nature of the ECM and the dualistic roles of its components across GI cancers, with a focus on their contributions to immune evasion and metabolic remodeling via intercellular interactions. Additionally, we explore therapeutic strategies targeting ECM remodeling and ECM-centered interactions, emphasizing their potential in enhancing existing anti-tumor therapies.
Collapse
Affiliation(s)
- Jiyifan Li
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenxin Zhang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Lu Chen
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinhai Wang
- Department of Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiafeng Liu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuxin Huang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Huijie Qi
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Li Chen
- Department of Pharmacy, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, China
| | - Tianxiao Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China.
| | - Qunyi Li
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
22
|
Domonkos L, Yusenko M, Kovacs G, Banyai D. Partial regression of conventional renal cell carcinoma displays markers of wound repair. J Clin Pathol 2024:jcp-2024-209459. [PMID: 39433307 DOI: 10.1136/jcp-2024-209459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/05/2024] [Indexed: 10/23/2024]
Abstract
AIMS During detailed analysis of H&E-stained histological slides of 710 unbiased conventional renal cell carcinomas (cRCCs), 141 tumours displayed partial regressive changes showing strong similarity to that of wound healing. We aimed to analyse the molecular processes occurring in regressive tumours. METHODS Immunohistochemistry was applied to analyse the signalling molecules in 12 selected tumours, and statistical analysis was used to estimate the correlation between regression and the outcome of the disease. RESULTS The regressive areas displayed inflammatory granulation tissue expressing transforming growth factor beta-1 (TGFB1), interleukin-1 beta and interleukin-6 (IL1B and IL6), proliferation of alpha smooth muscle actin (αSMA) positive naïve activated fibroblasts and a diffuse fibronectin 1 (FN1) network. In the central areas of regressive tissues, parallel-running myofibroblasts showed FN1, collagen type I alpha 1 (COL1A1) and collagen type III alpha 1 (COL3A1) positive immunoreaction. Partial tumour regression is associated with a better postoperative course of the disease. CONCLUSIONS Partial regression is a frequent event in cRCCs. Recognising complex molecular processes involved in tumour regression might help to find a way towards 'healing' cRCC.
Collapse
Affiliation(s)
| | - Maria Yusenko
- Ruhr University Bochum, Bochum, Nordrhein-Westfalen, Germany
| | - Gyula Kovacs
- University of Pecs Medical School, Pecs, Hungary
| | | |
Collapse
|
23
|
Kesti E, Borgmästars E, Hagström J, Mustonen H, Seppänen H, Haglund C, Sund M. The Prognostic Significance of Collagen VI in Pancreatic Ductal Adenocarcinoma. Pancreas 2024; 53:e729-e738. [PMID: 38913551 DOI: 10.1097/mpa.0000000000002360] [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] [Indexed: 06/26/2024]
Abstract
OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) is characterized by poor prognosis and lack of biomarkers. A rich desmoplastic tumor stroma is considered a hallmark of PDAC and previous studies have indicated upregulated expression of collagen VI (COL6) in PDAC. COL6 is shown to associate with prognosis in many cancers but has been less extensively studied in PDAC. MATERIALS AND METHODS The expression of COL6 was analyzed by immunohistochemistry in tissue microarrays containing resected tumor tissue samples from PDAC patients (n = 164). Significance of COL6 was estimated with Kaplan-Meier survival estimates and multivariable Cox regression analysis. COL6 protein and mRNA expression patterns were further investigated in publicly available datasets. RESULTS There were no statistically significant ( P < 0.05) differences in survival when comparing high and low protein expression of any of the analyzed COL6 α-chains (α1(VI): hazard ratio [HR] 0.90, 95% confidence interval [CI] 0.64-1.28; α2(VI): HR 1.28, 95% CI 0.86-1.89; α3(VI): HR 0.91, 95% CI 0.64-1.29). Similar results were obtained when assessing public data from the Cancer Proteome Atlas, Clinical Proteomic Tumor Analysis Consortium, and The Cancer Genome Atlas. CONCLUSIONS In contrast with previous studies and some other cancers, we did not find any association of COL6 tissue expression and PDAC survival.
Collapse
Affiliation(s)
- Ella Kesti
- From the Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Emmy Borgmästars
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
| | | | | | | | | | | |
Collapse
|
24
|
Wang Y, Jia J, Wang F, Fang Y, Yang Y, Zhou Q, Yuan W, Gu X, Hu J, Yang S. Pre-metastatic niche: formation, characteristics and therapeutic implication. Signal Transduct Target Ther 2024; 9:236. [PMID: 39317708 PMCID: PMC11422510 DOI: 10.1038/s41392-024-01937-7] [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: 02/28/2024] [Revised: 06/29/2024] [Accepted: 07/23/2024] [Indexed: 09/26/2024] Open
Abstract
Distant metastasis is a primary cause of mortality and contributes to poor surgical outcomes in cancer patients. Before the development of organ-specific metastasis, the formation of a pre-metastatic niche is pivotal in promoting the spread of cancer cells. This review delves into the intricate landscape of the pre-metastatic niche, focusing on the roles of tumor-derived secreted factors, extracellular vesicles, and circulating tumor cells in shaping the metastatic niche. The discussion encompasses cellular elements such as macrophages, neutrophils, bone marrow-derived suppressive cells, and T/B cells, in addition to molecular factors like secreted substances from tumors and extracellular vesicles, within the framework of pre-metastatic niche formation. Insights into the temporal mechanisms of pre-metastatic niche formation such as epithelial-mesenchymal transition, immunosuppression, extracellular matrix remodeling, metabolic reprogramming, vascular permeability and angiogenesis are provided. Furthermore, the landscape of pre-metastatic niche in different metastatic organs like lymph nodes, lungs, liver, brain, and bones is elucidated. Therapeutic approaches targeting the cellular and molecular components of pre-metastatic niche, as well as interventions targeting signaling pathways such as the TGF-β, VEGF, and MET pathways, are highlighted. This review aims to enhance our understanding of pre-metastatic niche dynamics and provide insights for developing effective therapeutic strategies to combat tumor metastasis.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Jiachi Jia
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Fuqi Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Yingshuai Fang
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Yabing Yang
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Quanbo Zhou
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Xiaoming Gu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Junhong Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| |
Collapse
|
25
|
Zhao J, Wang H, Chen J, Wang C, Gong N, Zhou F, Li X, Cao Y, Zhang H, Wang W, Zheng H, Zhang C. An oncolytic HSV-1 armed with Visfatin enhances antitumor effects by remodeling tumor microenvironment against murine pancreatic cancer. Biochem Biophys Res Commun 2024; 718:149931. [PMID: 38723415 DOI: 10.1016/j.bbrc.2024.149931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 05/23/2024]
Abstract
Oncolytic viruses (OVs) have shown potential in converting a "cold" tumor into a "hot" one and exhibit effectiveness in various cancer types. However, only a subset of patients respond to oncolytic virotherapy. It is important to understand the resistance mechanisms to OV treatment in pancreatic ductal adenocarcinoma (PDAC) to engineer oncolytic viruses. In this study, we used transcriptome RNA sequencing (RNA-seq) to identify Visfatin, which was highly expressed in the responsive tumors following OV treatment. To explore the antitumor efficacy, we modified OV-mVisfatin, which effectively inhibited tumor growth. For the first time, we revealed that Visfatin promoted the antitumor efficacy of OV by remodeling the tumor microenvironment, which involved enhancing CD8+ T cell and DC cell infiltration and activation, repolarizing macrophages towards the M1-like phenotype, and decreasing Treg cells using single-cell RNA sequencing (scRNA-seq) and flow cytometry. Furthermore, PD-1 blockade significantly enhanced OV-mVisfatin antitumor efficacy, offering a promising new therapeutic strategy for PDAC.
Collapse
Affiliation(s)
- Jiliang Zhao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Han Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Jinhua Chen
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Chunlei Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Nanxin Gong
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Feilong Zhou
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Xin Li
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Youjia Cao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China; Nankai International Advanced Research Institute (Shenzhen Futian), Nankai University, Shenzhen, China
| | - Hongkai Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Wei Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Hao Zheng
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China
| | - Cuizhu Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, China; Nankai International Advanced Research Institute (Shenzhen Futian), Nankai University, Shenzhen, China.
| |
Collapse
|
26
|
Qu J, Yan Z, Lei D, Zhong T, Fang C, Wen Z, Liu J, Lai Z, Yu XF, Zheng B, Geng S. Effect of Bioactive Black Phosphorus Nanomaterials on Cancer-Associated Fibroblast Heterogeneity in Pancreatic Cancer. ACS NANO 2024; 18:19354-19368. [PMID: 38975953 DOI: 10.1021/acsnano.4c06147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Tumor-stromal interactions and stromal heterogeneity in the tumor microenvironment are critical factors that influence the progression, metastasis, and chemoresistance of pancreatic ductal adenocarcinoma (PDAC). Here, we used spatial transcriptome technology to profile the gene expression landscape of primary PDAC and liver metastatic PDAC after bioactive black phosphorus nanomaterial (bioactive BP) treatment using a murine model of PDAC (LSL-KrasG12D/+; LSL-Trp53R172H/+; and Pdx-1-Cre mice). Bioinformatic and biochemical analyses showed that bioactive BP contributes to the tumor-stromal interplay by suppressing cancer-associated fibroblast (CAF) activation. Our results showed that bioactive BP contributes to CAF heterogeneity by decreasing the amount of inflammatory CAFs and myofibroblastic CAFs, two CAF subpopulations. Our study demonstrates the influence of bioactive BP on tumor-stromal interactions and CAF heterogeneity and suggests bioactive BP as a potential PDAC treatment.
Collapse
Affiliation(s)
- Jianhua Qu
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Zilong Yan
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Defeng Lei
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Tongning Zhong
- Central Laboratory, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Chongzhou Fang
- Central Laboratory, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Zonghua Wen
- Department of Pathology, Shenzhen University General Hospital, Shenzhen University, Shenzhen 518055, China
| | - Jikui Liu
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Zhengquan Lai
- Department of Pharmacy, Shenzhen University General Hospital/Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen 518055, China
| | - Xue-Feng Yu
- Shenzhen Key Laboratory of Micro/Nano Biosensing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Biao Zheng
- Department of Surgery, The First Dongguan Affiliated Hospital, Guangdong Medical University, No. 42 Jiaoping Road, Tangxia Town, Dongguan 523710, China
| | - Shengyong Geng
- Shenzhen Key Laboratory of Micro/Nano Biosensing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
27
|
Han D, Yang M, Feng Z, Wu Y, Sojic N, Jiang D. Thickness-Resolved Electrochemiluminescence Microscopy of Extracellular Matrix at Tumor Tissues for Rapid Cancer Diagnosis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:32078-32086. [PMID: 38865735 DOI: 10.1021/acsami.4c05735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
The traditional recognition of extracellular matrix (ECM) at tissue sections relies on the time-consuming immunofluorescence that could not meet the demand of rapid diagnosis. Herein, we introduce a thickness-resolved electrochemiluminescence (ECL) microscopy to image thin-layer ECM at tissue sections for fast histopathological analysis. The unique surface-confined ECL mechanism enables to unveil the diversity and complexity of multiple tissue structures with varying thicknesses. Notably, the short lifetimes and the limited diffusion of electrogenerated coreactant radicals combined with their chemical reactivity result in a 2-fold increase in ECL intensity on ECM structures compared to the remaining tissue, enabling ECM visualization without specific labeling. The further quantitation of the ECM localization within tissue sections furnishes crucial insights into tumor progression and, more importantly, differentiates carcinoma and paracancerous tissues from patients in less than 30 min. Moreover, the reported electrochemistry-based microscopy is a dynamic approach allowing to investigate the transport, tortuosity, and trafficking properties through the tissues. This thickness-resolved recognition strategy not only opens new avenues for imaging complex samples but also holds promise for expediting tissue pathologic diagnosis, offering a more automated protocol with enhanced quantitative data compared to current intraoperative pathology methods.
Collapse
Affiliation(s)
- Dongni Han
- State Key Laboratory of Analytical Chemistry for Life Science and School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
| | - Mi Yang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and Clinical Cancer Institute of Nanjing University, Nanjing 210008, China
| | - Zengyu Feng
- Department of General Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yulian Wu
- Department of General Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Neso Sojic
- Institute des Sciences Moléculaires, UMR 5255, 16 avenue Pey-Berland, University of Bordeaux, Pessac 33607, France
| | - Dechen Jiang
- State Key Laboratory of Analytical Chemistry for Life Science and School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
| |
Collapse
|
28
|
Ba Q, Wang X, Hu H, Lu Y. Single-Cell RNA Sequencing Analysis Reveals Metabolic Changes in Epithelial Glycosphingolipids and Establishes a Prognostic Risk Model for Pancreatic Cancer. Diagnostics (Basel) 2024; 14:1094. [PMID: 38893622 PMCID: PMC11171987 DOI: 10.3390/diagnostics14111094] [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: 04/15/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
OBJECTIVE Metabolic reprogramming serves as a distinctive feature of cancer, impacting proliferation and metastasis, with aberrant glycosphingolipid expression playing a crucial role in malignancy. Nevertheless, limited research has investigated the connection between glycosphingolipid metabolism and pancreatic cancer. METHODS This study utilized a single-cell sequencing dataset to analyze the cell composition in pancreatic cancer tissues and quantified single-cell metabolism using a newly developed computational pipeline called scMetabolism. A gene signature developed from the differential expressed genes (DEGs), related to epithelial cell glycosphingolipid metabolism, was established to forecast patient survival, immune response, mutation status, and reaction to chemotherapy with pancreatic adenocarcinoma (PAAD). RESULTS The single-cell sequencing analysis revealed a significant increase in epithelial cell proportions in PAAD, with high glycosphingolipid metabolism occurring in the cancerous tissue. A six-gene signature prognostic model based on abnormal epithelial glycosphingolipid metabolism was created and confirmed using publicly available databases. Patients with PAAD were divided into high- and low-risk categories according to the median risk score, with those in the high-risk group demonstrating a more unfavorable survival outcome in all three cohorts, with higher rates of gene mutations (e.g., KRAS, CDKN2A), increased levels of immunosuppressive cells (macrophages, Th2 cells, regulatory T cells), and heightened sensitivity to Acetalax and Selumetinlb. CONCLUSIONS Abnormal metabolism of glycosphingolipids in epithelial cells may promote the development of PAAD. A model utilizing a gene signature associated with epithelial glycosphingolipids metabolism has been established, serving as a valuable indicator for the prognostic stratification of patients with PAAD.
Collapse
Affiliation(s)
| | | | | | - Yanjun Lu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
29
|
Lee DU, Han BS, Jung KH, Hong SS. Tumor Stroma as a Therapeutic Target for Pancreatic Ductal Adenocarcinoma. Biomol Ther (Seoul) 2024; 32:281-290. [PMID: 38590092 PMCID: PMC11063484 DOI: 10.4062/biomolther.2024.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/10/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis owing to its desmoplastic stroma. Therefore, therapeutic strategies targeting this tumor stroma should be developed. In this study, we describe the heterogeneity of cancer-associated fibroblasts (CAFs) and their diverse roles in the progression, immune evasion, and resistance to treatment of PDAC. We subclassified the spatial distribution and functional activity of CAFs to highlight their effects on prognosis and drug delivery. Extracellular matrix components such as collagen and hyaluronan are described for their roles in tumor behavior and treatment outcomes, implying their potential as therapeutic targets. We also discussed the roles of extracellular matrix (ECM) including matrix metalloproteinases and tissue inhibitors in PDAC progression. Finally, we explored the role of the adaptive and innate immune systems in shaping the PDAC microenvironment and potential therapeutic strategies, with a focus on immune cell subsets, cytokines, and immunosuppressive mechanisms. These insights provide a comprehensive understanding of PDAC and pave the way for the development of prognostic markers and therapeutic interventions.
Collapse
Affiliation(s)
- Dae Ui Lee
- Department of Medicine, College of Medicine, Inha University, Incheon 22332, Republic of Korea
| | - Beom Seok Han
- Program in Biomedical Science & Engineering, The Graduate School, Inha University, Incheon 22212, Republic of Korea
| | - Kyung Hee Jung
- Department of Medicine, College of Medicine, Inha University, Incheon 22332, Republic of Korea
| | - Soon-Sun Hong
- Department of Medicine, College of Medicine, Inha University, Incheon 22332, Republic of Korea
- Program in Biomedical Science & Engineering, The Graduate School, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
30
|
Randall J, Hunt AL, Nutcharoen A, Johnston L, Chouraichi S, Wang H, Winer A, Wadlow R, Huynh J, Davis J, Corgiat B, Bateman NW, Deeken JF, Petricoin EF, Conrads TP, Cannon TL. Quantitative proteomic analysis of HER2 protein expression in PDAC tumors. Clin Proteomics 2024; 21:24. [PMID: 38509475 PMCID: PMC10953162 DOI: 10.1186/s12014-024-09476-7] [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: 11/16/2023] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Metastatic pancreatic adenocarcinoma (PDAC) is the third leading cause of cancer-related death in the United States, with a 5-year survival rate of only 11%, necessitating identification of novel treatment paradigms. Tumor tissue specimens from patients with PDAC, breast cancer, and other solid tumor malignancies were collected and tumor cells were enriched using laser microdissection (LMD). Reverse phase protein array (RPPA) analysis was performed on enriched tumor cell lysates to quantify a 32-protein/phosphoprotein biomarker panel comprising known anticancer drug targets and/or cancer-related total and phosphorylated proteins, including HER2Total, HER2Y1248, and HER3Y1289. RPPA analysis revealed significant levels of HER2Total in PDAC patients at abundances comparable to HER2-positive (IHC 3+) and HER2-low (IHC 1+ /2+ , FISH-) breast cancer tissues, for which HER2 screening is routinely performed. These data support a critical unmet need for routine clinical evaluation of HER2 expression in PDAC patients and examination of the utility of HER2-directed antibody-drug conjugates in these patients.
Collapse
Affiliation(s)
- Jamie Randall
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Dr, Fairfax, VA, 22031, USA
| | - Allison L Hunt
- Women's Health Integrated Research Center, Women's Service Line, Inova Health System, 3289 Woodburn Rd, Annandale, VA, 22042, USA
- Gynecologic Cancer Center of Excellence, Gynecologic Surgery and Obstetrics, Uniformed Services University and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
| | - Aratara Nutcharoen
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Dr, Fairfax, VA, 22031, USA
- Department of Pathology, Inova Fairfax Hospital, 3300 Gallows Road, Falls Church, VA, 22042, USA
| | - Laura Johnston
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Dr, Fairfax, VA, 22031, USA
| | - Safae Chouraichi
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Dr, Fairfax, VA, 22031, USA
| | - Hongkun Wang
- Department of Biostatistics, Bioinformatics, and Biomathematics, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Arthur Winer
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Dr, Fairfax, VA, 22031, USA
| | - Raymond Wadlow
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Dr, Fairfax, VA, 22031, USA
| | - Jasmine Huynh
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Dr, Fairfax, VA, 22031, USA
| | - Justin Davis
- Theralink Technologies, Inc., 15000 W 6th Ave, Golden, CO, 80401, USA
| | - Brian Corgiat
- Theralink Technologies, Inc., 15000 W 6th Ave, Golden, CO, 80401, USA
| | - Nicholas W Bateman
- Gynecologic Cancer Center of Excellence, Gynecologic Surgery and Obstetrics, Uniformed Services University and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - John F Deeken
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Dr, Fairfax, VA, 22031, USA
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 20110, USA
| | - Thomas P Conrads
- Women's Health Integrated Research Center, Women's Service Line, Inova Health System, 3289 Woodburn Rd, Annandale, VA, 22042, USA
- Gynecologic Cancer Center of Excellence, Gynecologic Surgery and Obstetrics, Uniformed Services University and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
| | - Timothy L Cannon
- Inova Schar Cancer Institute, Inova Health System, 8081 Innovation Park Dr, Fairfax, VA, 22031, USA.
| |
Collapse
|
31
|
Zhen L, Zhu Y, Wu Z, Liao J, Deng L, Ma Q, Wu Q, Ning G, Lin Q, Zhou L, Huang Y, Zhuo Z, Chen R, Yu D. Activated hedgehog gene pattern correlates with dismal clinical outcome and tumor microenvironment heterogeneity in hepatocellular carcinoma. Heliyon 2024; 10:e26989. [PMID: 38468970 PMCID: PMC10926087 DOI: 10.1016/j.heliyon.2024.e26989] [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: 05/07/2023] [Revised: 02/02/2024] [Accepted: 02/22/2024] [Indexed: 03/13/2024] Open
Abstract
Background Activation of the Hedgehog signaling pathway is linked to the initiation and development of human hepatocellular carcinoma (HCC). However, its impact on clinical outcomes and the HCC microenvironment remains unclear. Methods We performed comprehensive analyses of Hedgehog pathway genes in a large cohort of HCC patients. Specifically, we utilized univariate Cox regression analysis to identify Hedgehog genes linked to overall survival, and the LASSO algorithm was used to construct a Hedgehog-related gene pattern. We subsequently examined the correlation between the Hedgehog pattern and the HCC microenvironment employing the CIBERSORT and ssGSEA algorithms. Furthermore, Tumor Immune Dysfunction and Exclusion (TIDE) algorithm and the anti-PD-L1 treatment dataset (IMvigor210) are used to evaluate the clinical response of the Hedgehog pattern in predicting immune checkpoint inhibitors. Results We found that the Hedgehog activation score (HHAS), a prognostic score based on 11 Hedgehog genes, was significantly associated with HCC patient survival. Patients exhibiting high HHAS experienced markedly reduced survival rates compared to those with low HHAS, and HHAS emerged as an independent prognostic factor for HCC. Functional enrichment analysis unveiled the association of the HHAS phenotype with functions related to the immune system, and further investigation demonstrated that HCC patients exhibiting low HHAS displayed elevated levels of anti-tumor immune activation in CD8+ T cells, while high HHAS were linked to immune escape phenotypes and increased infiltration of immune suppressive cells. In addition, in the Immune Checkpoint Inhibitor (ICI) cohort of IMvigor210, patients with higher HHAS had worse ICI treatment outcomes and shortened survival time, indicating that the HHAS is a useful indicator for predicting patient response to immunotherapy. Conclusions In summary, our study offers valuable insights for advancing research on Hedgehog and its impact on tumor immunity, which provides an opportunity to optimize prognosis and immune therapy for HCC.
Collapse
Affiliation(s)
- Limin Zhen
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
- Department of Infectious Diseases, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Yi Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
- Department of Anesthesiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Zhen Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
- Department of Infectious Diseases, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Jinyao Liao
- Department of Infectious Diseases, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Liaoyuan Deng
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Qianqian Ma
- Department of Infectious Diseases, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Qili Wu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Gang Ning
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Diseases Center, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Qiuxiong Lin
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Liya Zhou
- Department of Radiation Oncology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Yanjie Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Zewei Zhuo
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Ren Chen
- Department of Infectious Diseases, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Dongnan Yu
- Department of Anesthesiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| |
Collapse
|
32
|
Xiao C, Li J, Hua A, Wang X, Li S, Li Z, Xu C, Zhang Z, Yang X, Li Z. Hyperbaric Oxygen Boosts Antitumor Efficacy of Copper-Diethyldithiocarbamate Nanoparticles against Pancreatic Ductal Adenocarcinoma by Regulating Cancer Stem Cell Metabolism. RESEARCH (WASHINGTON, D.C.) 2024; 7:0335. [PMID: 38766644 PMCID: PMC11100349 DOI: 10.34133/research.0335] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 02/16/2024] [Indexed: 05/22/2024]
Abstract
Cuproptosis-based cancer nanomedicine has received widespread attention recently. However, cuproptosis nanomedicine against pancreatic ductal adenocarcinoma (PDAC) is severely limited by cancer stem cells (CSCs), which reside in the hypoxic stroma and adopt glycolysis metabolism accordingly to resist cuproptosis-induced mitochondria damage. Here, we leverage hyperbaric oxygen (HBO) to regulate CSC metabolism by overcoming tumor hypoxia and to augment CSC elimination efficacy of polydopamine and hydroxyethyl starch stabilized copper-diethyldithiocarbamate nanoparticles (CuET@PH NPs). Mechanistically, while HBO and CuET@PH NPs inhibit glycolysis and oxidative phosphorylation, respectively, the combination of HBO and CuET@PH NPs potently suppresses energy metabolism of CSCs, thereby achieving robust tumor inhibition of PDAC and elongating mice survival importantly. This study reveals novel insights into the effects of cuproptosis nanomedicine on PDAC CSC metabolism and suggests that the combination of HBO with cuproptosis nanomedicine holds significant clinical translation potential for PDAC patients.
Collapse
Affiliation(s)
- Chen Xiao
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Jiayuan Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Ao Hua
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Xing Wang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Shiyou Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Zheng Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Chen Xu
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Zhijie Zhang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Xiangliang Yang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- National Engineering Research Center for Nanomedicine,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of Education,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Bioinformatics and Molecular Imaging Key Laboratory,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Zifu Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- National Engineering Research Center for Nanomedicine,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of Education,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
- Hubei Bioinformatics and Molecular Imaging Key Laboratory,
Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| |
Collapse
|
33
|
Zhou H, Li R, Liu J, Long J, Chen T. Characterization and verification of CAF-relevant prognostic gene signature to aid therapy in bladder cancer. Heliyon 2024; 10:e23873. [PMID: 38317915 PMCID: PMC10839800 DOI: 10.1016/j.heliyon.2023.e23873] [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: 04/18/2023] [Revised: 11/30/2023] [Accepted: 12/14/2023] [Indexed: 02/07/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) are significantly involved in determining the patient's prognosis and response to bladder cancer (BLCA) therapy. CAFs can induce epithelial-mesenchymal transformation (EMT) as well as complex interaction with immune cells. Hence, it is imperative to identify potential markers for enhancing our understanding of CAFs in BLCA progression and immune regulation. A variety of algorithms and analyses were employed in the study, leading to the development of a novel prognostic feature for CAFs-Stromal-EMT (CSE)-prognostic feature. This feature was constructed based on the genes MFAP5, PCOLCE2, and JUN. Furthermore, we revealed that patients with higher CSE risk scores responded to immunotherapy better compared to those with lower. Finally, we verified two CSE-related genes using in vitro experiments. Our results suggested that the CSE-prognostic feature could predict the prognosis and evaluate the response of patients to immune and chemotherapies. This would aid clinicians in designing treatment strategies for patients with BLCA.
Collapse
Affiliation(s)
- Huidong Zhou
- Department of Urology, Changsha Hospital of Hunan Normal University, Changsha, China
| | - Ruqi Li
- Department of Electrocardiography, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Jinghong Liu
- Department of Urology, Changsha Hospital of Hunan Normal University, Changsha, China
| | - Jianhua Long
- Department of Urology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Tao Chen
- Department of Urology, Changsha Hospital of Hunan Normal University, Changsha, China
| |
Collapse
|
34
|
Philipp LM, Yesilyurt UU, Surrow A, Künstner A, Mehdorn AS, Hauser C, Gundlach JP, Will O, Hoffmann P, Stahmer L, Franzenburg S, Knaack H, Schumacher U, Busch H, Sebens S. Epithelial and Mesenchymal-like Pancreatic Cancer Cells Exhibit Different Stem Cell Phenotypes Associated with Different Metastatic Propensities. Cancers (Basel) 2024; 16:686. [PMID: 38398077 PMCID: PMC10886860 DOI: 10.3390/cancers16040686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is mostly diagnosed at advanced or even metastasized stages, limiting the prognoses of patients. Metastasis requires high tumor cell plasticity, implying phenotypic switching in response to changing environments. Here, epithelial-mesenchymal transition (EMT), being associated with an increase in cancer stem cell (CSC) properties, and its reversion are important. Since it is poorly understood whether different CSC phenotypes exist along the EMT axis and how these impact malignancy-associated properties, we aimed to characterize CSC populations of epithelial and mesenchymal-like PDAC cells. Single-cell cloning revealed CSC (Holoclone) and non-CSC (Paraclone) clones from the PDAC cell lines Panc1 and Panc89. The Panc1 Holoclone cells showed a mesenchymal-like phenotype, dominated by a high expression of the stemness marker Nestin, while the Panc89 Holoclone cells exhibited a SOX2-dominated epithelial phenotype. The Panc89 Holoclone cells showed enhanced cell growth and a self-renewal capacity but slow cluster-like invasion. Contrarily, the Panc1 Holoclone cells showed slower cell growth and self-renewal ability but were highly invasive. Moreover, cell variants differentially responded to chemotherapy. In vivo, the Panc1 and Panc89 cell variants significantly differed regarding the number and size of metastases, as well as organ manifestation, leading to different survival outcomes. Overall, these data support the existence of different CSC phenotypes along the EMT axis in PDAC, manifesting different metastatic propensities.
Collapse
Affiliation(s)
- Lisa-Marie Philipp
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Umut-Ulas Yesilyurt
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Arne Surrow
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23538 Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany
| | - Anne-Sophie Mehdorn
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Charlotte Hauser
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Jan-Paul Gundlach
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Olga Will
- Molecular Imaging North Competence Center, Clinic of Radiology and Neuroradiology, Kiel University, UKSH, Campus Kiel, 24118 Kiel, Germany
| | - Patrick Hoffmann
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Lea Stahmer
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Sören Franzenburg
- Institute of Clinical Molecular Biology, Kiel University, 24118 Kiel, Germany
| | - Hendrike Knaack
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
- Academic Affairs Office, Hannover Medical School, 30625 Hannover, Germany
| | - Udo Schumacher
- Department of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23538 Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany
| | - Susanne Sebens
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| |
Collapse
|
35
|
Uehara M, Domoto T, Takenaka S, Takeuchi O, Shimasaki T, Miyashita T, Minamoto T. Glycogen synthase kinase 3β: the nexus of chemoresistance, invasive capacity, and cancer stemness in pancreatic cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:4. [PMID: 38318525 PMCID: PMC10838383 DOI: 10.20517/cdr.2023.84] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/20/2023] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
The treatment of pancreatic cancer remains a significant clinical challenge due to the limited number of patients eligible for curative (R0) surgery, failures in the clinical development of targeted and immune therapies, and the pervasive acquisition of chemotherapeutic resistance. Refractory pancreatic cancer is typified by high invasiveness and resistance to therapy, with both attributes related to tumor cell stemness. These malignant characteristics mutually enhance each other, leading to rapid cancer progression. Over the past two decades, numerous studies have produced evidence of the pivotal role of glycogen synthase kinase (GSK)3β in the progression of over 25 different cancer types, including pancreatic cancer. In this review, we synthesize the current knowledge on the pathological roles of aberrant GSK3β in supporting tumor cell proliferation and invasion, as well as its contribution to gemcitabine resistance in pancreatic cancer. Importantly, we discuss the central role of GSK3β as a molecular hub that mechanistically connects chemoresistance, tumor cell invasion, and stemness in pancreatic cancer. We also discuss the involvement of GSK3β in the formation of desmoplastic tumor stroma and in promoting anti-cancer immune evasion, both of which constitute major obstacles to successful cancer treatment. Overall, GSK3β has characteristics of a promising therapeutic target to overcome chemoresistance in pancreatic cancer.
Collapse
Affiliation(s)
- Masahiro Uehara
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Authors contributed equally
| | - Takahiro Domoto
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Authors contributed equally
| | - Satoshi Takenaka
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
- Department of Surgery, Toyama City Hospital, Toyama 939-8511, Japan
| | - Osamu Takeuchi
- Biomedical Laboratory, Department of Research, Kitasato University Kitasato Institute Hospital, Tokyo 108-8642, Japan
| | - Takeo Shimasaki
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Japan
| | - Tomoharu Miyashita
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8641, Japan
- Department of Surgery, Toyama City Hospital, Toyama 939-8511, Japan
| | - Toshinari Minamoto
- Division of Translational and Clinical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
| |
Collapse
|
36
|
Orlacchio A, Muzyka S, Gonda TA. Epigenetic therapeutic strategies in pancreatic cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 383:1-40. [PMID: 38359967 DOI: 10.1016/bs.ircmb.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid malignancies, characterized by its aggressiveness and metastatic potential, with a 5-year survival rate of only 8-11%. Despite significant improvements in PDAC treatment and management, therapeutic alternatives are still limited. One of the main reasons is its high degree of intra- and inter-individual tumor heterogeneity which is established and maintained through a complex network of transcription factors and epigenetic regulators. Epigenetic drugs, have shown promising preclinical results in PDAC and are currently being evaluated in clinical trials both for their ability to sensitize cancer cells to cytotoxic drugs and to counteract the immunosuppressive characteristic of PDAC tumor microenvironment. In this review, we discuss the current status of epigenetic treatment strategies to overcome molecular and cellular PDAC heterogeneity in order to improve response to therapy.
Collapse
Affiliation(s)
- Arturo Orlacchio
- Division of Gastroenterology and Hepatology, New York University, New York, NY, United States
| | - Stephen Muzyka
- Division of Gastroenterology and Hepatology, New York University, New York, NY, United States
| | - Tamas A Gonda
- Division of Gastroenterology and Hepatology, New York University, New York, NY, United States.
| |
Collapse
|
37
|
Hunt AL, Bateman NW, Barakat W, Makohon-Moore SC, Abulez T, Driscoll JA, Schaaf JP, Hood BL, Conrads KA, Zhou M, Calvert V, Pierobon M, Loffredo J, Wilson KN, Litzi TJ, Teng PN, Oliver J, Mitchell D, Gist G, Rojas C, Blanton B, Darcy KM, Rao UNM, Petricoin EF, Phippen NT, Maxwell GL, Conrads TP. Mapping three-dimensional intratumor proteomic heterogeneity in uterine serous carcinoma by multiregion microsampling. Clin Proteomics 2024; 21:4. [PMID: 38254014 PMCID: PMC10804562 DOI: 10.1186/s12014-024-09451-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Although uterine serous carcinoma (USC) represents a small proportion of all uterine cancer cases, patients with this aggressive subtype typically have high rates of chemotherapy resistance and disease recurrence that collectively result in a disproportionately high death rate. The goal of this study was to provide a deeper view of the tumor microenvironment of this poorly characterized uterine cancer variant through multi-region microsampling and quantitative proteomics. METHODS Tumor epithelium, tumor-involved stroma, and whole "bulk" tissue were harvested by laser microdissection (LMD) from spatially resolved levels from nine USC patient tumor specimens and underwent proteomic analysis by mass spectrometry and reverse phase protein arrays, as well as transcriptomic analysis by RNA-sequencing for one patient's tumor. RESULTS LMD enriched cell subpopulations demonstrated varying degrees of relatedness, indicating substantial intratumor heterogeneity emphasizing the necessity for enrichment of cellular subpopulations prior to molecular analysis. Known prognostic biomarkers were quantified with stable levels in both LMD enriched tumor and stroma, which were shown to be highly variable in bulk tissue. These USC data were further used in a comparative analysis with a data generated from another serous gynecologic malignancy, high grade serous ovarian carcinoma, and have been added to our publicly available data analysis tool, the Heterogeneity Analysis Portal ( https://lmdomics.org/ ). CONCLUSIONS Here we identified extensive three-dimensional heterogeneity within the USC tumor microenvironment, with disease-relevant biomarkers present in both the tumor and the stroma. These data underscore the critical need for upfront enrichment of cellular subpopulations from tissue specimens for spatial proteogenomic analysis.
Collapse
Grants
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
- HU0001-16-2-0006, HU0001-19-2-0031, HU0001-20-2-0033, and HU0001-21-2-0027, and HU0001-22-2-0016 Defense Health Agency
Collapse
Affiliation(s)
- Allison L Hunt
- Women's Health Integrated Research Center, Inova Women's Service Line, Inova Health System, 3289 Woodburn Rd, Suite 375, Annandale, VA, 22042, USA
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
| | - Nicholas W Bateman
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
- Department of Surgery, The John P. Murtha Cancer Center Research Program, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
| | - Waleed Barakat
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Sasha C Makohon-Moore
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Tamara Abulez
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Jordan A Driscoll
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Joshua P Schaaf
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Brian L Hood
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Kelly A Conrads
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Ming Zhou
- Women's Health Integrated Research Center, Inova Women's Service Line, Inova Health System, 3289 Woodburn Rd, Suite 375, Annandale, VA, 22042, USA
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
| | - Valerie Calvert
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Mariaelena Pierobon
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Jeremy Loffredo
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Katlin N Wilson
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Tracy J Litzi
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Pang-Ning Teng
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Julie Oliver
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Dave Mitchell
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Glenn Gist
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Christine Rojas
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
| | - Brian Blanton
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Kathleen M Darcy
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
- Department of Surgery, The John P. Murtha Cancer Center Research Program, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
| | - Uma N M Rao
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Neil T Phippen
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Drive, Suite 100, Bethesda, MD, 20817, USA
- Department of Surgery, The John P. Murtha Cancer Center Research Program, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
| | - G Larry Maxwell
- Women's Health Integrated Research Center, Inova Women's Service Line, Inova Health System, 3289 Woodburn Rd, Suite 375, Annandale, VA, 22042, USA
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- Department of Surgery, The John P. Murtha Cancer Center Research Program, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
| | - Thomas P Conrads
- Women's Health Integrated Research Center, Inova Women's Service Line, Inova Health System, 3289 Woodburn Rd, Suite 375, Annandale, VA, 22042, USA.
- Gynecologic Cancer Center of Excellence and the Women's Health Integrated Research Center, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA.
- Department of Surgery, The John P. Murtha Cancer Center Research Program, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA.
| |
Collapse
|
38
|
Xu W, Cai J, Peng T, Meng T, Pang Y, Sun L, Wu H, Zhang J, Chen X, Chen H. Fibroblast Activation Protein-Targeted PET/CT with 18F-Fibroblast Activation Protein Inhibitor-74 for Evaluation of Gastrointestinal Cancer: Comparison with 18F-FDG PET/CT. J Nucl Med 2024; 65:40-51. [PMID: 37884330 DOI: 10.2967/jnumed.123.266329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
Fibroblast activation protein is overexpressed in the stroma of several cancer types. 18F-fibroblast activation protein inhibitor (FAPI)-74 is a PET tracer with high selectivity for fibroblast activation protein and has shown high accumulation in human tumors in clinical studies. However, the use of 18F-FAPI-74 for PET imaging of gastrointestinal cancer has not been systematically investigated. Herein, we investigated the diagnostic accuracy of 18F-FAPI-74 (18F-LNC1005) PET/CT in gastric, liver, and pancreatic cancers and compared the results with those of 18F-FDG PET/CT. Methods: This prospective study analyzed patients with confirmed gastric, liver, or pancreatic malignancies who underwent concurrent 18F-FDG and 18F-FAPI-74 PET/CT between June 2022 and December 2022. PET/CT findings were confirmed by histopathology or radiographic follow-up. 18F-FDG and 18F-FAPI-74 uptake and tumor-to-background ratios were compared using the Wilcoxon signed-rank test. The McNemar test was used to compare the diagnostic accuracy of the 2 scans. Results: Our cohort consisted of 112 patients: 49 with gastric cancer, 39 with liver cancer, and 24 with pancreatic cancer. Among them, 69 patients underwent PET/CT for initial staging and 43 for recurrence detection. Regarding lesion-based diagnostic accuracy, 18F-FAPI-74 PET/CT showed higher sensitivity than did 18F-FDG in the detection of primary tumors (gastric cancer, 88% [22/25] vs. 60% [15/25], P = 0.016; liver cancer, 100% [22/22] vs. 82% [18/22], P = 0.125; pancreatic cancer, 100% [22/22] vs. 86% [19/22], P = 0.250), local recurrence (92% [23/25] vs. 56% [14/25]; P = 0.021), involved lymph nodes (71% [41/58] vs. 40% [23/58]; P < 0.001), and bone and visceral metastases (98% [350/358] vs. 47% [168/358]; P < 0.001). Compared with 18F-FDG, 18F-FAPI-74 PET/CT upstaged 17 patients' TNM staging among all treatment-naïve patients (17/69, 25%) and changed the clinical management of 4 patients (4/43, 9%) in whom recurrence or metastases were detected. Conclusion: 18F-FAPI-74 PET/CT is superior to 18F-FDG PET/CT in detecting primary tumors, local recurrence, lymph node involvement, and bone and visceral metastases in gastric, pancreatic, and liver cancers, with higher uptake in most primary and metastatic lesions.
Collapse
Affiliation(s)
- Weizhi Xu
- Department of Nuclear Medicine, First Affiliated Hospital of Xiamen University, Xiamen, China
- Minnan PET Center, First Affiliated Hospital of Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Radiopharmaceuticals, School of Medicine, Xiamen University, Xiamen, China
| | - Jiayu Cai
- Department of Nuclear Medicine, First Affiliated Hospital of Xiamen University, Xiamen, China
- Minnan PET Center, First Affiliated Hospital of Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Radiopharmaceuticals, School of Medicine, Xiamen University, Xiamen, China
| | - Tianxing Peng
- Department of Nuclear Medicine, First Affiliated Hospital of Xiamen University, Xiamen, China
- Minnan PET Center, First Affiliated Hospital of Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Radiopharmaceuticals, School of Medicine, Xiamen University, Xiamen, China
| | - Tinghua Meng
- Department of Nuclear Medicine, First Affiliated Hospital of Xiamen University, Xiamen, China
- Minnan PET Center, First Affiliated Hospital of Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Radiopharmaceuticals, School of Medicine, Xiamen University, Xiamen, China
| | - Yizhen Pang
- Department of Nuclear Medicine, First Affiliated Hospital of Xiamen University, Xiamen, China
- Minnan PET Center, First Affiliated Hospital of Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Radiopharmaceuticals, School of Medicine, Xiamen University, Xiamen, China
| | - Long Sun
- Department of Nuclear Medicine, First Affiliated Hospital of Xiamen University, Xiamen, China
- Minnan PET Center, First Affiliated Hospital of Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Radiopharmaceuticals, School of Medicine, Xiamen University, Xiamen, China
| | - Hua Wu
- Department of Nuclear Medicine, First Affiliated Hospital of Xiamen University, Xiamen, China
- Minnan PET Center, First Affiliated Hospital of Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Radiopharmaceuticals, School of Medicine, Xiamen University, Xiamen, China
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Chemical and Biomolecular Engineering, College of Design and Engineering, National University of Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore; and
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore;
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Chemical and Biomolecular Engineering, College of Design and Engineering, National University of Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Haojun Chen
- Department of Nuclear Medicine, First Affiliated Hospital of Xiamen University, Xiamen, China;
- Minnan PET Center, First Affiliated Hospital of Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Radiopharmaceuticals, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
39
|
Ando R, Shiraki Y, Miyai Y, Shimizu H, Furuhashi K, Minatoguchi S, Kato K, Kato A, Iida T, Mizutani Y, Ito K, Asai N, Mii S, Esaki N, Takahashi M, Enomoto A. Meflin is a marker of pancreatic stellate cells involved in fibrosis and epithelial regeneration in the pancreas. J Pathol 2024; 262:61-75. [PMID: 37796386 DOI: 10.1002/path.6211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 07/18/2023] [Accepted: 08/25/2023] [Indexed: 10/06/2023]
Abstract
Pancreatic stellate cells (PSCs) are stromal cells in the pancreas that play an important role in pancreatic pathology. In chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC), PSCs are known to get activated to form myofibroblasts or cancer-associated fibroblasts (CAFs) that promote stromal fibroinflammatory reactions. However, previous studies on PSCs were mainly based on the findings obtained using ex vivo expanded PSCs, with few studies that addressed the significance of in situ tissue-resident PSCs using animal models. Their contributions to fibrotic reactions in CP and PDAC are also lesser-known. These limitations in our understanding of PSC biology have been attributed to the lack of specific molecular markers of PSCs. Herein, we established Meflin (Islr), a glycosylphosphatidylinositol-anchored membrane protein, as a PSC-specific marker in both mouse and human by using human pancreatic tissue samples and Meflin reporter mice. Meflin-positive (Meflin+ ) cells contain lipid droplets and express the conventional PSC marker Desmin in normal mouse pancreas, with some cells also positive for Gli1, the marker of pancreatic tissue-resident fibroblasts. Three-dimensional analysis of the cleared pancreas of Meflin reporter mice showed that Meflin+ PSCs have long and thin cytoplasmic protrusions, and are localised on the abluminal side of vessels in the normal pancreas. Lineage tracing experiments revealed that Meflin+ PSCs constitute one of the origins of fibroblasts and CAFs in CP and PDAC, respectively. In these diseases, Meflin+ PSC-derived fibroblasts showed a distinctive morphology and distribution from Meflin+ PSCs in the normal pancreas. Furthermore, we showed that the genetic depletion of Meflin+ PSCs accelerated fibrosis and attenuated epithelial regeneration and stromal R-spondin 3 expression, thereby implying that Meflin+ PSCs and their lineage cells may support tissue recovery and Wnt/R-spondin signalling after pancreatic injury and PDAC development. Together, these data indicate that Meflin may be a marker specific to tissue-resident PSCs and useful for studying their biology in both health and disease. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Ryota Ando
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihiro Shiraki
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Miyai
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Shimizu
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuhiro Furuhashi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shun Minatoguchi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Katsuhiro Kato
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akira Kato
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tadashi Iida
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuyuki Mizutani
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kisuke Ito
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoya Asai
- Department of Molecular Pathology, Fujita Health University, Toyoake, Japan
| | - Shinji Mii
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobutoshi Esaki
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahide Takahashi
- Division of International Center for Cell and Gene Therapy, Fujita Health University, Toyoake, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Gifu, Japan
| |
Collapse
|
40
|
Zhang T, Gu Z, Ni R, Wang X, Jiang Q, Tao R. An Update on Gemcitabine-Based Chemosensitization Strategies in Pancreatic Ductal Adenocarcinoma. FRONT BIOSCI-LANDMRK 2023; 28:361. [PMID: 38179740 DOI: 10.31083/j.fbl2812361] [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: 06/01/2023] [Revised: 07/22/2023] [Accepted: 08/10/2023] [Indexed: 01/06/2024]
Abstract
Pancreatic cancer is the seventh leading cause of cancer-related deaths, and chemotherapy is one of the most important treatments for pancreatic cancer. Unfortunately, pancreatic cancer cells can block chemotherapy drugs from entering the tumor. This is owing to interactions between the tumor's environment and the cancer cells. Here, we review the latest research on the mechanisms by which pancreatic cancer cells block the chemotherapy drug, gemcitabine. The results of our review can help identify potential therapeutic targets for the blocking of gemcitabine by pancreatic cancer cells and may provide new strategies to help chemotherapy drugs penetrate tumors.
Collapse
Affiliation(s)
- Tianpeng Zhang
- Department of Surgery, Bengbu Medical College, 233030 Bengbu, AnHui, China
| | - Zongting Gu
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 310000 Hangzhou, Zhejiang, China
| | - Ran Ni
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 310000 Hangzhou, Zhejiang, China
| | - Xiao Wang
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 310000 Hangzhou, Zhejiang, China
| | - Qitao Jiang
- Department of Surgery, Bengbu Medical College, 233030 Bengbu, AnHui, China
| | - Ran Tao
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 310000 Hangzhou, Zhejiang, China
| |
Collapse
|
41
|
Liang G, Oh TG, Hah N, Tiriac H, Shi Y, Truitt ML, Antal CE, Atkins AR, Li Y, Fraser C, Ng S, Pinto AFM, Nelson DC, Estepa G, Bashi S, Banayo E, Dai Y, Liddle C, Yu RT, Hunter T, Engle DD, Han H, Von Hoff DD, Downes M, Evans RM. Inhibiting stromal Class I HDACs curbs pancreatic cancer progression. Nat Commun 2023; 14:7791. [PMID: 38057326 PMCID: PMC10700526 DOI: 10.1038/s41467-023-42178-6] [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: 04/13/2023] [Accepted: 09/27/2023] [Indexed: 12/08/2023] Open
Abstract
Oncogenic lesions in pancreatic ductal adenocarcinoma (PDAC) hijack the epigenetic machinery in stromal components to establish a desmoplastic and therapeutic resistant tumor microenvironment (TME). Here we identify Class I histone deacetylases (HDACs) as key epigenetic factors facilitating the induction of pro-desmoplastic and pro-tumorigenic transcriptional programs in pancreatic stromal fibroblasts. Mechanistically, HDAC-mediated changes in chromatin architecture enable the activation of pro-desmoplastic programs directed by serum response factor (SRF) and forkhead box M1 (FOXM1). HDACs also coordinate fibroblast pro-inflammatory programs inducing leukemia inhibitory factor (LIF) expression, supporting paracrine pro-tumorigenic crosstalk. HDAC depletion in cancer-associated fibroblasts (CAFs) and treatment with the HDAC inhibitor entinostat (Ent) in PDAC mouse models reduce stromal activation and curb tumor progression. Notably, HDAC inhibition (HDACi) enriches a lipogenic fibroblast subpopulation, a potential precursor for myofibroblasts in the PDAC stroma. Overall, our study reveals the stromal targeting potential of HDACi, highlighting the utility of this epigenetic modulating approach in PDAC therapeutics.
Collapse
Affiliation(s)
- Gaoyang Liang
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Tae Gyu Oh
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Department of Oncology Science, OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA
| | - Nasun Hah
- Next Generation Sequencing Core, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Hervé Tiriac
- Department of Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yu Shi
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Bristol Myer Squibb, 10300 Campus Point Drive, Suite 100, San Diego, CA, 92121, USA
| | - Morgan L Truitt
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Corina E Antal
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Annette R Atkins
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Yuwenbin Li
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Cory Fraser
- HonorHealth Scottsdale Osborn Medical Center and Shea Medical Center, Scottsdale, AZ, 85260, USA
| | - Serina Ng
- Molecular Medicine Division, The Translational Genomic Research Institute, Phoenix, AZ, 85004, USA
| | - Antonio F M Pinto
- Mass Spectrometry Core, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Dylan C Nelson
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Gabriela Estepa
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Senada Bashi
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Ester Banayo
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Yang Dai
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Christopher Liddle
- Storr Liver Centre, Westmead Institute for Medical Research and Sydney Medical School, University of Sydney, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Ruth T Yu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Dannielle D Engle
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Haiyong Han
- Molecular Medicine Division, The Translational Genomic Research Institute, Phoenix, AZ, 85004, USA
| | - Daniel D Von Hoff
- HonorHealth Scottsdale Osborn Medical Center and Shea Medical Center, Scottsdale, AZ, 85260, USA
- Molecular Medicine Division, The Translational Genomic Research Institute, Phoenix, AZ, 85004, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
| |
Collapse
|
42
|
Chen G, Liu Y, Su D, Qiu J, Long J, Zhao F, Tao J, Yang G, Huang H, Xiao J, Zhang T, Zhao Y. Genomic analysis and filtration of novel prognostic biomarkers based on metabolic and immune subtypes in pancreatic cancer. Cell Oncol (Dordr) 2023; 46:1691-1708. [PMID: 37434012 DOI: 10.1007/s13402-023-00836-3] [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] [Accepted: 06/01/2023] [Indexed: 07/13/2023] Open
Abstract
PURPOSE Patients with pancreatic cancer (PC) can be classified into various molecular subtypes and benefit from some precise therapy. Nevertheless, the interaction between metabolic and immune subtypes in the tumor microenvironment (TME) remains unknown. We hope to identify molecular subtypes related to metabolism and immunity in pancreatic cancer METHODS: Unsupervised consensus clustering and ssGSEA analysis were utilized to construct molecular subtypes related to metabolism and immunity. Diverse metabolic and immune subtypes were characterized by distinct prognoses and TME. Afterward, we filtrated the overlapped genes based on the differentially expressed genes (DEGs) between the metabolic and immune subtypes by lasso regression and Cox regression, and used them to build risk score signature which led to PC patients was categorized into high- and low-risk groups. Nomogram were built to predict the survival rates of each PC patient. RT-PCR, in vitro cell proliferation assay, PC organoid, immunohistochemistry staining were used to identify key oncogenes related to PC RESULTS: High-risk patients have a better response for various chemotherapeutic drugs in the Genomics of Drug Sensitivity in Cancer (GDSC) database. We built a nomogram with the risk group, age, and the number of positive lymph nodes to predict the survival rates of each PC patient with average 1-year, 2-year, and 3-year areas under the curve (AUCs) equal to 0.792, 0.752, and 0.751. FAM83A, KLF5, LIPH, MYEOV were up-regulated in the PC cell line and PC tissues. Knockdown of FAM83A, KLF5, LIPH, MYEOV could reduce the proliferation in the PC cell line and PC organoids CONCLUSION: The risk score signature based on the metabolism and immune molecular subtypes can accurately predict the prognosis and guide treatments of PC, meanwhile, the metabolism-immune biomarkers may provide novel target therapy for PC.
Collapse
Affiliation(s)
- Guangyu Chen
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Department of Breast Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Yueze Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Dan Su
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Jiangdong Qiu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Junyu Long
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fangyu Zhao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Jinxin Tao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Gang Yang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Hua Huang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Jianchun Xiao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Taiping Zhang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China.
| | - Yupei Zhao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China.
| |
Collapse
|
43
|
Karbhari A, Mosessian S, Trivedi KH, Valla F, Jacobson M, Truty MJ, Patnam NG, Simeone DM, Zan E, Brennan T, Chen H, Kuo PH, Herrmann K, Goenka AH. Gallium-68-labeled fibroblast activation protein inhibitor-46 PET in patients with resectable or borderline resectable pancreatic ductal adenocarcinoma: A phase 2, multicenter, single arm, open label non-randomized study protocol. PLoS One 2023; 18:e0294564. [PMID: 38011131 PMCID: PMC10681241 DOI: 10.1371/journal.pone.0294564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/20/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease prone to widespread metastatic dissemination and characterized by a desmoplastic stroma that contributes to poor outcomes. Fibroblast activation protein (FAP)-expressing Cancer-Associated Fibroblasts (CAFs) are crucial components of the tumor stroma, influencing carcinogenesis, fibrosis, tumor growth, metastases, and treatment resistance. Non-invasive tools to profile CAF identity and function are essential for overcoming CAF-mediated therapy resistance, developing innovative targeted therapies, and improved patient outcomes. We present the design of a multicenter phase 2 study (clinicaltrials.gov identifier NCT05262855) of [68Ga]FAPI-46 PET to image FAP-expressing CAFs in resectable or borderline resectable PDAC. METHODS We will enroll up to 60 adult treatment-naïve patients with confirmed PDAC. These patients will be eligible for curative surgical resection, either without prior treatment (Cohort 1) or after neoadjuvant therapy (NAT) (Cohort 2). A baseline PET scan will be conducted from the vertex to mid-thighs approximately 15 minutes after administering 5 mCi (±2) of [68Ga]FAPI-46 intravenously. Cohort 2 patients will undergo an additional PET after completing NAT but before surgery. Histopathology and FAP immunohistochemistry (IHC) of initial diagnostic biopsy and resected tumor samples will serve as the truth standards. Primary objective is to assess the sensitivity, specificity, and accuracy of [68Ga]FAPI-46 PET for detecting FAP-expressing CAFs. Secondary objectives will assess predictive values and safety profile validation. Exploratory objectives are comparison of diagnostic performance of [68Ga]FAPI-46 PET to standard-of-care imaging, and comparison of pre- versus post-NAT [68Ga]FAPI-46 PET in Cohort 2. CONCLUSION To facilitate the clinical translation of [68Ga]FAPI-46 in PDAC, the current study seeks to implement a coherent strategy to mitigate risks and increase the probability of meeting FDA requirements and stakeholder expectations. The findings from this study could potentially serve as a foundation for a New Drug Application to the FDA. TRIAL REGISTRATION @ClinicalTrials.gov identifier NCT05262855.
Collapse
Affiliation(s)
- Aashna Karbhari
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Sherly Mosessian
- Clinical Development, Sofie Biosciences, Dulles, Virginia, United States of America
| | - Kamaxi H. Trivedi
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Frank Valla
- Radiopharmaceutical and Contract Manufacturing, Sofie Biosciences, Dulles, Virginia, United States of America
| | - Mark Jacobson
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Mark J. Truty
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Nandakumar G. Patnam
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Diane M. Simeone
- Departments of Surgery and Pathology, NYU Langone Health, New York, New York, United States of America
| | - Elcin Zan
- Department of Radiology, Weill Cornell Medicine, New York, New York, United States of America
| | - Tracy Brennan
- Discovery Life Sciences, Newtown, Pennsylvania, United States of America
| | - Hongli Chen
- Discovery Life Sciences, Newtown, Pennsylvania, United States of America
| | - Phillip H. Kuo
- Departments of Medical Imaging, Medicine and Biomedical Engineering, University of Arizona, Tucson, Arizona, United States of America
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ajit H. Goenka
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
44
|
Aguirre GA, Goulart MR, Dalli J, Kocher HM. Arachidonate 15-lipoxygenase-mediated production of Resolvin D5 n-3 DPA abrogates pancreatic stellate cell-induced cancer cell invasion. Front Immunol 2023; 14:1248547. [PMID: 38035115 PMCID: PMC10687150 DOI: 10.3389/fimmu.2023.1248547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023] Open
Abstract
Activation of pancreatic stellate cells (PSCs) to cancer-associated fibroblasts (CAFs) is responsible for the extensive desmoplastic reaction observed in PDAC stroma: a key driver of pancreatic ductal adenocarcinoma (PDAC) chemoresistance leading to poor prognosis. Specialized pro-resolving mediators (SPMs) are prime modulators of inflammation and its resolution, traditionally thought to be produced by immune cells. Using liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based lipid mediator profiling PSCs as well as primary human CAFs express enzymes and receptors to produce and respond to SPMs. Human PSC/CAF SPM secretion profile can be modulated by rendering these cells activated [transforming growth factor beta (TGF-β)] or quiescent [all-trans retinoic acid (ATRA)]. ATRA-induced nuclear translocation of arachidonate-15-lipoxygenase (ALOX15) was linked to increased production of n-3 docosapentaenoic acid-derived Resolvin D5 (RvD5n-3 DPA), among other SPMs. Inhibition of RvD5n-3 DPA formation increases cancer cell invasion, whereas addback of this molecule reduced activated PSC-mediated cancer cell invasion. We also observed that circulating concentrations of RvD5n-3 DPA levels were decreased in peripheral blood of metastatic PDAC patients when compared with those measured in plasma of non-metastatic PDAC patients. Together, these findings indicate that RvD5n-3 DPA may regulate cancer-stroma cross-talk and invasion.
Collapse
Affiliation(s)
- Gabriel A. Aguirre
- Centre for Tumour Biology, Barts Cancer Institute, London, United Kingdom
| | | | | | - Jesmond Dalli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, John Vane Science Centre, Queen Mary University of London, London, United Kingdom
| | - Hemant M. Kocher
- Centre for Tumour Biology, Barts Cancer Institute, London, United Kingdom
| |
Collapse
|
45
|
Du HX, Wang H, Ma XP, Chen H, Dai AB, Zhu KX. Eukaryotic translation initiation factor 2α kinase 2 in pancreatic cancer: An approach towards managing clinical prognosis and molecular immunological characterization. Oncol Lett 2023; 26:478. [PMID: 37818134 PMCID: PMC10561166 DOI: 10.3892/ol.2023.14066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 07/14/2023] [Indexed: 10/12/2023] Open
Abstract
Most patients with pancreatic cancer are already in the late stages of the disease when they are diagnosed, and pancreatic cancer is a deadly disease with a poor prognosis. With the advancement of research, immunotherapy has become a new focus in the treatment of tumors. To the best of our knowledge, there is currently no reliable diagnostic or prognostic marker for pancreatic cancer; therefore, the present study investigated the potential of eukaryotic translation initiation factor 2α kinase 2 (EIF2AK2) as a predictive and diagnostic marker for pancreatic cancer. Immunohistochemical staining of clinical samples independently verified that EIF2AK2 expression was significantly higher in clinically operated pancreatic cancer tissues than in adjacent pancreatic tissues., and EIF2AK2 expression and differentially expressed genes (DEGs) were identified using downloadable RNA sequencing data from The Cancer Genome Atlas and Genomic Tumor Expression Atlas. In addition, Gene Ontology/Kyoto Encyclopedia of Genes and Genomes analyses and immune cell infiltration were used for functional enrichment analysis of EIF2AK2-associated DEGs. The clinical importance of EIF2AK2 was also determined using Kaplan-Meier survival, Cox regression and time-dependent survival receiver operating characteristic curve analyses, and a predictive nomogram model was generated. Finally, the functional role of EIF2AK2 was assessed in PANC-1 cells using a short hairpin RNA-EIF2AK2 knockdown approach, including CCK-8, wound healing assay, cell cycle and apoptosis assays. The findings suggested that EIF2AK2 may have potential as a diagnostic and prognostic biomarker for patients with pancreatic cancer. Furthermore, EIF2AK2 may provide a new therapeutic target for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Hao-Xuan Du
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Hu Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xiao-Peng Ma
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Hao Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Ai-Bin Dai
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Ke-Xiang Zhu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
46
|
Bararia A, Das A, Mitra S, Banerjee S, Chatterjee A, Sikdar N. Deoxyribonucleic acid methylation driven aberrations in pancreatic cancer-related pathways. World J Gastrointest Oncol 2023; 15:1505-1519. [PMID: 37746645 PMCID: PMC10514732 DOI: 10.4251/wjgo.v15.i9.1505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/29/2023] [Accepted: 08/01/2023] [Indexed: 09/13/2023] Open
Abstract
Pancreatic cancer (PanCa) presents a catastrophic disease with poor overall survival at advanced stages, with immediate requirement of new and effective treatment options. Besides genetic mutations, epigenetic dysregulation of signaling pathway-associated enriched genes are considered as novel therapeutic target. Mechanisms beneath the deoxyribonucleic acid methylation and its utility in developing of epi-drugs in PanCa are under trails. Combinations of epigenetic medicines with conventional cytotoxic treatments or targeted therapy are promising options to improving the dismal response and survival rate of PanCa patients. Recent studies have identified potentially valid pathways that support the prediction that future PanCa clinical trials will include vigorous testing of epigenomic therapies. Epigenetics thus promises to generate a significant amount of new knowledge of biological and medical importance. Our review could identify various components of epigenetic mechanisms known to be involved in the initiation and development of pancreatic ductal adenocarcinoma and related precancerous lesions, and novel pharmacological strategies that target these components could potentially lead to breakthroughs. We aim to highlight the possibilities that exist and the potential therapeutic interventions.
Collapse
Affiliation(s)
- Akash Bararia
- Human Genetics Unit, Indian Statistical Institute, Kolkata 700108, India
| | - Amlan Das
- Department of Biochemistry, Royal Global University, Assam 781035, India
| | - Sangeeta Mitra
- Department of Biochemistry and Biophysics, University of Kalyani, West Bengal 741235, India
| | - Sudeep Banerjee
- Department of Gastrointestinal Surgery, Tata Medical Center, Kolkata 700160, India
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun 248007, India
| | - Nilabja Sikdar
- Human Genetics Unit, Indian Statistical Institute, Kolkata 700108, India
| |
Collapse
|
47
|
Liang G, Oh TG, Hah N, Tiriac H, Shi Y, Truitt ML, Antal CE, Atkins AR, Li Y, Fraser C, Ng S, Pinto AFM, Nelson DC, Estepa G, Bashi S, Banayo E, Dai Y, Liddle C, Yu RT, Hunter T, Engle DD, Han H, Von Hoff DD, Downes M, Evans RM. Inhibiting Stromal Class I HDACs Curbs Pancreatic Cancer Progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.12.557260. [PMID: 37745372 PMCID: PMC10515810 DOI: 10.1101/2023.09.12.557260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Oncogenic lesions in pancreatic ductal adenocarcinoma (PDAC) hijack the epigenetic machinery in stromal components to establish a desmoplastic and therapeutic resistant tumor microenvironment (TME). Here we identify Class I histone deacetylases (HDACs) as key epigenetic factors facilitating the induction of pro-desmoplastic and pro-tumorigenic transcriptional programs in pancreatic stromal fibroblasts. Mechanistically, HDAC-mediated changes in chromatin architecture enable the activation of pro-desmoplastic programs directed by serum response factor (SRF) and forkhead box M1 (FOXM1). HDACs also coordinate fibroblast pro-inflammatory programs inducing leukemia inhibitory factor (LIF) expression, supporting paracrine pro-tumorigenic crosstalk. HDAC depletion in cancer-associated fibroblasts (CAFs) and treatment with the HDAC inhibitor entinostat (Ent) in PDAC mouse models reduce stromal activation and curb tumor progression. Notably, HDAC inhibition (HDACi) enriches a lipogenic fibroblast subpopulation, a potential precursor for myofibroblasts in the PDAC stroma. Overall, our study reveals the stromal targeting potential of HDACi, highlighting the utility of this epigenetic modulating approach in PDAC therapeutics.
Collapse
|
48
|
Chitty JL, Yam M, Perryman L, Parker AL, Skhinas JN, Setargew YFI, Mok ETY, Tran E, Grant RD, Latham SL, Pereira BA, Ritchie SC, Murphy KJ, Trpceski M, Findlay AD, Melenec P, Filipe EC, Nadalini A, Velayuthar S, Major G, Wyllie K, Papanicolaou M, Ratnaseelan S, Phillips PA, Sharbeen G, Youkhana J, Russo A, Blackwell A, Hastings JF, Lucas MC, Chambers CR, Reed DA, Stoehr J, Vennin C, Pidsley R, Zaratzian A, Da Silva AM, Tayao M, Charlton B, Herrmann D, Nobis M, Clark SJ, Biankin AV, Johns AL, Croucher DR, Nagrial A, Gill AJ, Grimmond SM, Pajic M, Timpson P, Jarolimek W, Cox TR. A first-in-class pan-lysyl oxidase inhibitor impairs stromal remodeling and enhances gemcitabine response and survival in pancreatic cancer. NATURE CANCER 2023; 4:1326-1344. [PMID: 37640930 PMCID: PMC10518255 DOI: 10.1038/s43018-023-00614-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 07/07/2023] [Indexed: 08/31/2023]
Abstract
The lysyl oxidase family represents a promising target in stromal targeting of solid tumors due to the importance of this family in crosslinking and stabilizing fibrillar collagens and its known role in tumor desmoplasia. Using small-molecule drug-design approaches, we generated and validated PXS-5505, a first-in-class highly selective and potent pan-lysyl oxidase inhibitor. We demonstrate in vitro and in vivo that pan-lysyl oxidase inhibition decreases chemotherapy-induced pancreatic tumor desmoplasia and stiffness, reduces cancer cell invasion and metastasis, improves tumor perfusion and enhances the efficacy of chemotherapy in the autochthonous genetically engineered KPC model, while also demonstrating antifibrotic effects in human patient-derived xenograft models of pancreatic cancer. PXS-5505 is orally bioavailable, safe and effective at inhibiting lysyl oxidase activity in tissues. Our findings present the rationale for progression of a pan-lysyl oxidase inhibitor aimed at eliciting a reduction in stromal matrix to potentiate chemotherapy in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Jessica L Chitty
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Michelle Yam
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Lara Perryman
- Pharmaxis, Frenchs Forest, New South Wales, Australia
| | - Amelia L Parker
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Joanna N Skhinas
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Yordanos F I Setargew
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Ellie T Y Mok
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Emmi Tran
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Rhiannon D Grant
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Sharissa L Latham
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Brooke A Pereira
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Shona C Ritchie
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Kendelle J Murphy
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Michael Trpceski
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | | | - Pauline Melenec
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Elysse C Filipe
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Audrey Nadalini
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Sipiththa Velayuthar
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Gretel Major
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Kaitlin Wyllie
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Michael Papanicolaou
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Shivanjali Ratnaseelan
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Phoebe A Phillips
- School of Biomedical Sciences, Faculty of Medicine, Lowy Cancer Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
| | - George Sharbeen
- School of Biomedical Sciences, Faculty of Medicine, Lowy Cancer Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
| | - Janet Youkhana
- School of Biomedical Sciences, Faculty of Medicine, Lowy Cancer Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
| | - Alice Russo
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Antonia Blackwell
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Jordan F Hastings
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Morghan C Lucas
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Cecilia R Chambers
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Daniel A Reed
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Janett Stoehr
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Claire Vennin
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Ruth Pidsley
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Anaiis Zaratzian
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Andrew M Da Silva
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Michael Tayao
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | | | - David Herrmann
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Max Nobis
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- Intravital Imaging Expertise Center, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Susan J Clark
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, UK
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
| | - Amber L Johns
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - David R Croucher
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Adnan Nagrial
- Department of Medical Oncology, Westmead Hospital, Sydney, New South Wales, Australia
| | - Anthony J Gill
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, New South Wales, Australia
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, Sydney, New South Wales, Australia
| | - Sean M Grimmond
- University of Melbourne Centre for Cancer Research, VCCC, Melbourne, Victoria, Australia
| | - Marina Pajic
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Paul Timpson
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | | | - Thomas R Cox
- Cancer Ecosystems Program, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, New South Wales, Australia.
- School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, UNSW Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
49
|
Byeon S, du Toit‐Thompson T, Gillson J, Gill AJ, Samra JS, Mittal A, Sahni S. Heterogeneous tumor microenvironment in pancreatic ductal adenocarcinoma: An emerging role of single-cell analysis. Cancer Med 2023; 12:18020-18031. [PMID: 37537839 PMCID: PMC10523961 DOI: 10.1002/cam4.6407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/21/2023] [Accepted: 07/25/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies in the world, for which the mortality is almost as high as the disease incidence and is predicted to be the second-highest cause of cancer-related deaths by 2030. These cancerous tumors consist of diversified gene expressions within the different cellular subpopulations that include neoplastic ductal cells, cancer-associated fibroblasts, and immune cells, all of which collectively facilitate cellular heterogeneity in the PDAC tumor microenvironment (TME). Active intratumoral interaction within the cell populations in TME induces the proliferation of cancerous cells, accounting for tumorigenesis and rapid metastasis. METHODS This review will focus on novel findings uncovering PDAC heterogeneity in different cellular subpopulations using single-cell RNA-sequencing (scRNA-seq) and other single-cell analysis technologies. It will further explore the emerging role of single-cell technologies in assessing the role of different subpopulations of neoplastic ductal cells, cancer-associated fibroblasts, and immune cells in PDAC progression. RESULTS AND CONCLUSION The application of scRNA-seq in PDAC has started to unveil associations between disease progression and heterogeneity in pancreatic TME and could influence future PDAC treatment. Recent advances in scRNA-seq have uncovered comprehensive analyses of heterogeneous ecosystems present within the TME. These emerging findings underpins further need for a more in-depth understanding of intratumoral heterogeneity in the PDAC microenvironment.
Collapse
Affiliation(s)
- Sooin Byeon
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
- Kolling Institute of Medical Research, University of SydneySydneyNew South WalesAustralia
| | - Taymin du Toit‐Thompson
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
- Kolling Institute of Medical Research, University of SydneySydneyNew South WalesAustralia
| | - Josef Gillson
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
- Kolling Institute of Medical Research, University of SydneySydneyNew South WalesAustralia
| | - Anthony J. Gill
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
- Kolling Institute of Medical Research, University of SydneySydneyNew South WalesAustralia
- Australian Pancreatic CentreSydneyNew South WalesAustralia
- Cancer Diagnosis and Pathology GroupKolling Institute of Medical ResearchSt LeonardsNew South WalesAustralia
- NSW Health Pathology, Department of Anatomical PathologyRoyal North Shore HospitalSt LeonardsNew South WalesAustralia
| | - Jaswinder S. Samra
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
- Australian Pancreatic CentreSydneyNew South WalesAustralia
- Upper GI Surgical UnitRoyal North Shore Hospital and North Shore Private HospitalSt LeonardsNew South WalesAustralia
| | - Anubhav Mittal
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
- Australian Pancreatic CentreSydneyNew South WalesAustralia
- Upper GI Surgical UnitRoyal North Shore Hospital and North Shore Private HospitalSt LeonardsNew South WalesAustralia
- The University of Notre Dame AustraliaSydneyNew South WalesAustralia
| | - Sumit Sahni
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
- Kolling Institute of Medical Research, University of SydneySydneyNew South WalesAustralia
- Australian Pancreatic CentreSydneyNew South WalesAustralia
| |
Collapse
|
50
|
Zhang C, Lei D, Zhou Y, Zhong T, Li X, Ai W, Zheng B, Liu J, Piao Y, Yan Z, Lai Z. Identifying a baicalein-related prognostic signature contributes to prognosis prediction and tumor microenvironment of pancreatic cancer. Front Immunol 2023; 14:1223650. [PMID: 37575248 PMCID: PMC10416623 DOI: 10.3389/fimmu.2023.1223650] [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: 05/23/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant and lethal human cancers in the world due to its high metastatic potential, and patients with PDAC have a poor prognosis, yet quite little is understood regarding the underlying biological mechanisms of its high metastatic capacity. Baicalein has a dramatic anti-tumor function in the treatment of different types of cancer. However, the therapeutic effects of baicalein on human PDAC and its mechanisms of action have not been extensively understood. In order to explore the biological characteristic, molecular mechanisms, and potential clinical value of baicalein in inhibiting the metastatic capacity of PDAC. We performed several in vitro, in vivo, and in silico studies. We first examined the potential regulation of baicalein in the metastatic capacity of PDAC cells. We showed that baicalein could dramatically suppress liver metastasis of PDAC cells with highly metastatic potential in mice model. The high-throughput sequencing analysis was employed to explore the biological roles of baicalein in PDAC cells. We found that baicalein might be involved in the infiltration of Cancer-Associated Fibroblasts (CAF) in PDAC. Moreover, a baicalein-related risk model and a lncRNA-related model were built by Cox analysis according to the data set of PDAC from TCGA database which suggested a clinical value of baicalein. Finally, we revealed a potential downstream target of baicalein in PDAC, we proposed that baicalein might contribute to the infiltration of CAF via FGFBP1. Thus, we uncovered a novel role for baicalein in regulation of PDAC liver metastasis that may contribute to its anti-cancer effect. We proposed that baicalein might suppress PDAC liver metastasis via regulation of FGFBP1-mediated CAF infiltration. Our results provide a new perspective on clinical utility of baicalein and open new avenues for the inhibition of liver-metastasis of PDAC.
Collapse
Affiliation(s)
- Citing Zhang
- Department of Pharmacy, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen, Guangdong, China
| | - Defeng Lei
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yan Zhou
- Department of Obstetrics & Carson International Cancer Research Center, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy, Shenzhen, Guangdong, China
| | - Tongning Zhong
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Xuefei Li
- College of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| | - Weipeng Ai
- Department of Pharmacy, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen, Guangdong, China
| | - Biao Zheng
- Department of Surgery, The First Dongguan Affiliated Hospital, Guangdong Medical University. Dongguan, Guangdong, China
| | - Jikui Liu
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yicui Piao
- Department of Critical Care Medicine, National Cancer Center, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, China
| | - Zilong Yan
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Zhengquan Lai
- Department of Pharmacy, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|