1
|
Lin D, Li Y, Fang J, Xie X, Zhang B, Ye X, Huang Y, Zhang X, Xue A. Global, regional and national burden of pancreatitis in children and adolescents, 1990-2021: a systematic analysis for the global burden of disease study 2021. Ann Med 2025; 57:2499699. [PMID: 40326502 PMCID: PMC12057782 DOI: 10.1080/07853890.2025.2499699] [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: 10/09/2024] [Revised: 03/08/2025] [Accepted: 03/18/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Pancreatitis poses a significant global health burden, disproportionately affecting children and adolescents. This study uses the global burden of disease (GBD) 2021 dataset to evaluate pancreatitis epidemiology in this demographic, focusing on disparities by age, sex, and region. OBJECTIVE To assess global trends in pediatric pancreatitis, identify risk factors, and forecast disease burden to 2035. METHODS We analysed GBD 2021 data on deaths and disability-adjusted life years (DALYs) for pancreatitis in individuals under 20. The socio-demographic index (SDI) assessed the link between societal development and health outcomes. Bayesian age-period-cohort (BAPC) modelling and Poisson's linear models were applied to project future burdens and estimate annual percentage changes (EAPCs) in age-standardized rates. RESULTS In 2021, pancreatitis caused 1120.09 deaths in children and adolescents, comprising 2% of all pancreatitis-related deaths. Age-standardized death rate (ASDR) and DALYs rate declined from 1990 to 2021 (EAPC -0.92 and -0.86, respectively). Low-middle SDI regions, notably Andean and Central Latin America and Eastern Europe, faced the highest burden. Alcohol was a leading risk factor, accounting for 3.51% of related deaths, and males had higher death and DALYs rate. CONCLUSIONS Despite declining pancreatitis-related mortality and DALYs, the disease remains a challenge, particularly in low-middle SDI regions. Alcohol consumption is a key risk factor, underscoring the need for targeted public health interventions. Gender-, age-, and region-specific strategies are essential to mitigate pancreatitis impact in children and adolescents.
Collapse
Affiliation(s)
- Derong Lin
- Dongguan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, China
| | - Yue Li
- Dongguan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, China
| | - Jingya Fang
- Dongguan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, China
| | - Xiaohua Xie
- Dongguan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, China
| | - Bin Zhang
- The Fourth Affiliated Hospital of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Xiaolin Ye
- The Seventh Affiliated Hospital of Guangzhou University of Chinese Medicine, Bao’an District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Yiheng Huang
- Dongguan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, China
| | - Xiaowen Zhang
- Dongguan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, China
| | - Aiguo Xue
- Dongguan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, China
| |
Collapse
|
2
|
Zhang P, Pan J, Lin S, Peng B, An C, Zhang J, Xu L, Lai Y, Yu H, Xu Z. Smart drug delivery platforms reprogramming cancer immune cycle to mitigate immune resistance of pancreatic tumors. Adv Drug Deliv Rev 2025; 224:115620. [DOI: 10.1016/j.addr.2025.115620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2025]
|
3
|
Zhang Z, Zhang Q, Wang Y. CAF-mediated tumor vascularization: From mechanistic insights to targeted therapies. Cell Signal 2025; 132:111827. [PMID: 40288665 DOI: 10.1016/j.cellsig.2025.111827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are a major component of the tumor microenvironment (TME) and play a crucial role in tumor progression. The biological properties of tumors, such as drug resistance, vascularization, immunosuppression, and metastasis are closely associated with CAFs. During tumor development, CAFs contribute to tumor progression by remodeling the extracellular matrix (ECM), inhibiting immune cell function, promoting angiogenesis, and facilitating tumor cell growth, invasion, and metastasis. Studies have shown that CAFs can promote endothelial cell proliferation by directly secreting cytokines such as vascular endothelial growth factor (VEGF) and fibroblast Growth Factor (FGF), as well as through exosomes. CAFs also secrete the chemokine stromal cell-derived factor 1 (SDF-1) to recruit endothelial progenitor cells (EPCs) into the peripheral blood and guide their migration to the tumor periphery. Additionally, CAFs can induce tumor cells to transform into "endothelial cells" that participate in vascular wall formation. However, the precise mechanisms remain to be further investigated. Due to their widespread presence in various solid tumors and their tumor-promoting function, CAFs are emerging as therapeutic targets. In this review, we summarize the specific mechanisms through which CAFs promote angiogenesis and outline current therapeutic strategies targeting CAF-induced vascularization, ongoing clinical trials targeting CAFs, and discuss potential future treatment approaches. We hope this will contribute to the advancement of CAF-targeted tumor treatment strategies.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Qing Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yang Wang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
4
|
Malhotra P, Fyfe J, Emmanouilidi A, Casari I, Mellett NA, Huynh K, Pajic M, Greening DW, Meikle PJ, Falasca M. Oncogenic small extracellular vesicles enriched in sphingosine-1-phosphate play a crucial role in pancreatic cancer progression. Cell Signal 2025; 132:111775. [PMID: 40158707 DOI: 10.1016/j.cellsig.2025.111775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Small extracellular vesicles (sEVs) from tumour cells mediate intercellular communication and signalling to regulate the progression of pancreatic ductal adenocarcinoma (PDAC). While we and others have shown that PDAC-derived sEVs comprise oncogenic protein and nucleic acid cargo, understanding the lipid landscape of these sEVs remains unknown. Lipids influence both the composition of sEVs and their roles in lipid metabolism and signalling pathways within the tumour microenvironment and tumorigenesis. We hypothesised that specific lipids in oncogenic sEVs might provide insights into PDAC. Comprehensive mass spectrometry-based lipidomic analysis was performed using liquid chromatography-electrospray ionisation-tandem mass spectrometry on sEVs isolated from PDAC and non-malignant pancreatic cell lines, patient-derived xenograft cell lines and plasma from the PDAC transgenic mouse model KPC (KRASWT/G12D/ TP53WT/R172H/Pdx1-Cre+/+). The sEV lipidomic analyses identified over 700 lipid species from 25 lipid classes and subclasses. Our results showed that, compared to non-malignant cells, PDAC-derived sEVs were enriched in specific lysophospholipids, particularly sphingosine-1-phosphate (S1P), a lipid known for its pivotal role in cancer pathogenesis. S1P enrichment was validated in plasma-derived sEVs from KPC mice compared to WT. To explore the functional implications of S1P enrichment, we conducted assays demonstrating that S1P in sEVs facilitated tubule formation in human microvascular endothelial cells and promoted cancer-associated fibroblast cell migration. We show that PDAC-derived sEVs are differentially enriched in specific lipids associated with cancer phenotype. Our findings highlight that PDAC-derived sEVs are enriched in specific lipids, particularly S1P, which plays a crucial role in promoting cancer progression.
Collapse
Affiliation(s)
- Pratibha Malhotra
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Jordan Fyfe
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Aikaterini Emmanouilidi
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Ilaria Casari
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Natalie A Mellett
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Kevin Huynh
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Marina Pajic
- Translational Oncology Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - David W Greening
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia; Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia; School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Marco Falasca
- University of Parma, Department of Medicine and Surgery, Via Volturno 39, 43125 Parma, Italy.
| |
Collapse
|
5
|
Zhang H, Ma T, Wen X, Jiang J, Chen J, Jiang J, Xie J, Mo T, Li R, Xie H, Liang G, Wang L, Zheng Z, Huang X, Liu C, Baihetiyaer Y, Abulimiti A, He X, Chen Z, Hu T, Pan W. SIK1 promotes ferroptosis resistance in pancreatic cancer via HDAC5-STAT6-SLC7A11 axis. Cancer Lett 2025; 623:217726. [PMID: 40250791 DOI: 10.1016/j.canlet.2025.217726] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
The activation of protein kinases is ubiquitous in pancreatic ductal adenocarcinoma (PDAC), yet its impact on ferroptosis remains unclear. SIK1 was identified as a key regulator of ferroptosis resistance in PDAC by kinase database screening. Targeting SIK1 could significantly reverse ferroptosis resistance and enhance cytotoxic effects of gemcitabine via increasing ferroptosis sensitivity in PDAC cells. Mechanistically, SIK1 phosphorylated HDAC5 at Ser498 residue and promoted its interaction with 14-3-3 protein, which further protected HDAC5 from TRIM28-mediated ubiquitylation and degradation. SIK1-stabilized HDAC5 deacetylated STAT6 and enhanced its transcriptional activity to upregulate SLC7A11 expression, ultimately rendering PDAC cells resistance to ferroptosis. SIK1 inhibitor (YKL-05-099) could synergistically enhance the antitumor effects of gemcitabine in organoid and patient-derived xenograft (PDX) models by inducing ferroptosis, suggesting a novel therapeutic target for PDAC. Clinically, SIK1 was positively correlated with SLC7A11 expression in PDAC specimens, which was associated with poor prognosis. These findings unveil a crucial mechanism through which PDAC counters ferroptosis via SIK1-mediated HDAC5 stabilization and subsequent SLC7A11 upregulation. This study underscores the promising potential of targeting SIK1-HDAC5 axis as a therapeutic strategy to overcome drug resistance in PDAC.
Collapse
Affiliation(s)
- Hao Zhang
- Department of General Surgery (Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Ma
- Department of General Surgery (Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaofeng Wen
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianlong Jiang
- Department of Gastrointestinal Surgery, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Jing Chen
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junfeng Jiang
- Department of General Surgery (Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiancong Xie
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Taiwei Mo
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ruibing Li
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hanlin Xie
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guanzhan Liang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lin Wang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zheyu Zheng
- Department of General Surgery (Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoming Huang
- Department of General Surgery (Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chuanyuan Liu
- Department of General Surgery, The Ganzhou People's Hospital, Ganzhou, China
| | - Yimamu Baihetiyaer
- Department of General Surgery, The First People's Hospital of Kashgar Region, Kashgar, China
| | - Abuduhalike Abulimiti
- Department of General Surgery, The Second People's Hospital of Kashgar Region, Kashgar, China
| | - Xiaosheng He
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Zexian Chen
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Tuo Hu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Weidong Pan
- Department of General Surgery (Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
6
|
Blanke EN, Holmes GM. Dysfunction of pancreatic exocrine secretion after experimental spinal cord injury. Exp Neurol 2025; 389:115257. [PMID: 40221007 PMCID: PMC12063635 DOI: 10.1016/j.expneurol.2025.115257] [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: 01/08/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
Pancreatic exocrine dysfunction is an underdiagnosed comorbidity in individuals living with spinal cord injury (SCI) who often present cholestasis, acute pancreatitis or high levels of serum pancreatic enzymes. Parasympathetic control of pancreatic exocrine secretion (PES) is mediated in the medullary dorsal vagal complex in part through cholecystokinin (CCK) release. Our previous reports indicate high thoracic (T3-) SCI reduces vagal afferent sensitivity to GI regulatory peptides, like CCK and thyrotropin releasing hormone (TRH). To date, the effects of experimental SCI on PES are unknown. Here we investigated the modulation of PES following T3-SCI in rats. We measured PES volume and amylase concentration in control and T3-SCI rats (3-days or 3-weeks after injury) following: (i) intra-duodenal administration of a mixed-nutrient liquid meal (Ensure® ™) or (ii) central TRH injection (100 pmol) in the dorsal motor nucleus of the vagus. In a separate cohort of overnight-fasted rats, basal serum amylase levels were measured. The baseline volume of PES secretion was lower in 3-week rats destined to receive Ensure® or TRH following T3-SCI surgery compared to control. PES protein concentration was significantly reduced at baseline in 3-week T3-SCI and elevated in 3-day and 3-week T3-SCI rats postprandially but only elevated in 3-day rats following TRH microinjection. Serum amylase activity levels were elevated in 3-day T3-SCI rats and remained at similar levels post 3-weeks T3-SCI. Our data suggest that vagally-mediated regulation of multiple visceral organs is disrupted in the days and weeks following experimental SCI.
Collapse
Affiliation(s)
- Emily N Blanke
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States of America; Department of Biology, Pennsylvania State University, York, PA 17403, United States of America
| | - Gregory M Holmes
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States of America.
| |
Collapse
|
7
|
Liu L, Zhang Y, Liu H, Yang J, Tian Q, Chueakula N, Ramasamy S, Verma NK, Cheung C, Kusumbe AP. Transforming Cancer Therapy: Unlocking the Potential of Targeting Vascular and Stromal Cells in the Tumor Microenvironment. Cancer Res 2025; 85:2152-2164. [PMID: 40173050 DOI: 10.1158/0008-5472.can-24-4744] [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: 12/11/2024] [Revised: 01/27/2025] [Accepted: 03/31/2025] [Indexed: 04/04/2025]
Abstract
The tumor microenvironment (TME) orchestrates cancer progression by fostering a complex interplay between cancer cells and the surrounding cellular and acellular elements. Through dynamic interactions with cancer cells, vascular and stromal cells not only promote tumor growth but also enhance metastatic potential and restrict therapeutic responses. Vascular and stromal cells play a critical role in regulating epithelial-mesenchymal transition (EMT) and sustaining resistance pathways, making them compelling targets for innovative therapies. This review delves into the vascular and stromal components of the TME, their contributions to EMT and resistance mechanisms, and emerging strategies to target these interactions for improved cancer therapy outcomes.
Collapse
Affiliation(s)
- Lu Liu
- Tissue and Tumor Microenvironments Lab, Cancer Discovery and Regenerative Medicine Program, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Yuheng Zhang
- Tissue and Tumor Microenvironments Lab, Cancer Discovery and Regenerative Medicine Program, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Hanyu Liu
- Tissue and Tumor Microenvironments Lab, Cancer Discovery and Regenerative Medicine Program, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jian Yang
- Tissue and Tumor Microenvironments Lab, Cancer Discovery and Regenerative Medicine Program, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Qi Tian
- Tissue and Tumor Microenvironments Lab, Cancer Discovery and Regenerative Medicine Program, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Multidisciplinary Institute of Ageing (MIA-Portugal), Coimbra, Portugal
| | - Nareekarn Chueakula
- Tissue and Tumor Microenvironments Lab, Cancer Discovery and Regenerative Medicine Program, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Saravana Ramasamy
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironments Lab, Cancer Discovery and Regenerative Medicine Program, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Multidisciplinary Institute of Ageing (MIA-Portugal), Coimbra, Portugal
| |
Collapse
|
8
|
Du Nguyen D, Shuklin F, Barulina E, Albitskaya H, Novikov S, Chernov AI, Kim I, Barulin A. Recent advances in dynamic single-molecule analysis platforms for diagnostics: Advantages over bulk assays and miniaturization approaches. Biosens Bioelectron 2025; 278:117361. [PMID: 40117897 DOI: 10.1016/j.bios.2025.117361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/23/2025]
Abstract
Single-molecule science is a unique technique for unraveling molecular biophysical processes. Sensitivity to single molecules provides the capacity for the early diagnosis of low biomarker amounts. Furthermore, the miniaturization of instruments for portable diagnostic tools toward point-of-care testing (POCT) is a crucial development in this field. Herein, we discuss recent developments in single-molecule sensing platforms and their advantages for diagnostics over bulk measurements including molecular size measurements, interaction dynamics, and fast biomarker sensing and sequencing at low concentrations. We highlight the capabilities of dynamic optical and electrical sensing platforms for single-biomolecule and single-vesicle monitoring associated with neurodegenerative disorders, viral diseases, cancers, and more. Current approaches to instrument miniaturization have brought technology closer to portable diagnostics settings via smartphone-based devices, multifunctional portable microscopes, handheld electrical circuit devices, and remote single-molecule assays. Finally, we provide an overview of the clinical applications of single-molecule sensors in POCT assays. Altogether, single-molecule analyses platforms exhibit significant potential for the development of novel portable healthcare devices.
Collapse
Affiliation(s)
- Dang Du Nguyen
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Fedor Shuklin
- Moscow Center for Advanced Studies, Kulakova str. 20, Moscow, 123592, Russia
| | - Elena Barulina
- Moscow Center for Advanced Studies, Kulakova str. 20, Moscow, 123592, Russia; Russian Quantum Center, Moscow, 121205, Russia
| | - Hristina Albitskaya
- Moscow Center for Advanced Studies, Kulakova str. 20, Moscow, 123592, Russia
| | - Sergey Novikov
- Moscow Center for Advanced Studies, Kulakova str. 20, Moscow, 123592, Russia
| | - Alexander I Chernov
- Russian Quantum Center, Moscow, 121205, Russia; Center for Photonics and 2D Materials, Moscow Institute of Physics and Technology, Dolgoprudny, 141700, Russia.
| | - Inki Kim
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Department of MetaBioHealth, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Aleksandr Barulin
- Moscow Center for Advanced Studies, Kulakova str. 20, Moscow, 123592, Russia.
| |
Collapse
|
9
|
Lee SI, Nam AR, Oh KS, Kim JM, Bang JH, Jeong Y, Choo SY, Kim HJ, Yoon J, Kim TY, Oh DY. Therapeutic potential of BOLD-100, a GRP78 inhibitor, enhanced by ATR inhibition in pancreatic ductal adenocarcinoma. Cell Commun Signal 2025; 23:281. [PMID: 40514666 PMCID: PMC12164152 DOI: 10.1186/s12964-025-02242-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 05/09/2025] [Indexed: 06/16/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by poor prognosis and resistance to conventional therapies, necessitating novel treatments. The high proliferative rate and protein synthesis in PDAC induce endoplasmic reticulum (ER) stress, with Glucose-Regulated Protein 78 (GRP78), a key regulator of ER stress and the Unfolded Protein Response (UPR), playing a pivotal role in PDAC progression. Despite its relevance, GRP78-targeted therapies remain unexplored in PDAC. BOLD-100, a novel GRP78 inhibitor, presents a potential therapeutic approach by disrupting GRP78 transcription, though its effects on PDAC have yet to be fully elucidated. Here, we found that BOLD-100 induces PDAC cell death through the UPR pathway activation, leading to CHOP-dependent apoptosis. BOLD-100 generates reactive oxygen species (ROS), inducing R-loop formation that triggers a DNA damage response via the ATR/Chk1 axis. BOLD-100 synergizes with AZD6738, an ATR inhibitor, to enhance anti-tumor efficacy compared to either agent alone in both in vitro and in vivo models. These findings suggest that BOLD-100, especially in combination with an ATR inhibitor, represents a promising therapeutic option for patients with PDAC.
Collapse
Affiliation(s)
- Su In Lee
- Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
| | - Ah-Rong Nam
- Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
| | - Kyoung-Seok Oh
- Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
| | - Jae-Min Kim
- Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, 03080, Korea
| | - Ju-Hee Bang
- Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
| | - Yoojin Jeong
- Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
| | - Sea Young Choo
- Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
| | - Hyo Jung Kim
- Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
| | - Jeesun Yoon
- Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Korea
| | - Tae-Yong Kim
- Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Korea
| | - Do-Youn Oh
- Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea.
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, 03080, Korea.
- Department of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Korea.
| |
Collapse
|
10
|
Chen S, Zhao Y, Cao M, Peng W, Huang H, Yang Y, Liang J, Chen W, Bai S, Zhou Q, Jiang J, Gu Y, Wang R, Cheng B. Anti-CTGF/PD-1 bispecific antibody Y126S restrains desmoplastic and immunosuppressive microenvironment in pancreatic cancer. J Immunother Cancer 2025; 13:e012144. [PMID: 40514068 DOI: 10.1136/jitc-2025-012144] [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] [Accepted: 05/29/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is characterized by a desmoplastic and immunosuppressive tumor microenvironment (TME), limiting the efficacy of immune checkpoint inhibitors such as anti-programmed cell death 1 (PD-1). METHODS This study aimed to evaluate the therapeutic potential of Y126S, a recombinant IgG1/IgG2 hybrid bispecific antibody (BsAb), in reshaping the immunotherapy-resistant TME in PDAC. Orthotopic PDAC and KPC (KrasLSL-G12D/+; Trp53LSL-R172H/+; Pdx1-Cre) mouse models were established and treated with Y126S, α-connective tissue growth factor (CTGF), α-PD-1, or a combination of α-CTGF and α-PD-1. TME remodeling, antibody distribution, and therapeutic efficacy were assessed using flow cytometry, immunohistochemical/Masson staining, atomic force microscopy, positron emission tomography (PET) imaging, distribution analysis, and other experimental techniques. RESULTS Here, Y126S was characterized in vitro and its antitumor efficacy was evaluated and validated in orthotopic PDAC mice and KPC mouse models. Notably, Y126S significantly remodeled the TME and demonstrated superior tumor-specific accumulation compared with single α-PD-1 treatment, leading to markedly enhanced antitumor efficacy relative to its parental antibodies or their combination. Mechanistically, Y126S suppressed cancer-associated fibroblasts (CAFs) activation, reduced collagen deposition, and downregulated programmed cell death ligand 1 (PD-L1) expression on CAFs by targeting CTGF and enhanced the anti-PD-1-mediated reinvigoration of cytotoxic CD8+ T cells, thereby establishing a less desmoplastic and potent tumor-killing microenvironment. CONCLUSIONS Our findings highlight the potential of Y126S as a promising BsAb-based immunotherapy strategy for PDAC by remodeling the desmoplastic and immunosuppressive TME.
Collapse
Affiliation(s)
- Shiru Chen
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuchong Zhao
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mengdie Cao
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wang Peng
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yilei Yang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jingwen Liang
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Chen
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Shuya Bai
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiaodan Zhou
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiamei Jiang
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yilin Gu
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ronghua Wang
- Department of Internal Medicine, University of Pittsburgh Medical Center Mercy Hospital, Pittsburgh, Pennsylvania, USA
| | - Bin Cheng
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
11
|
Zhou C, Geng J, Wu Z, Dan L, Huang H, Ruan X, Chen J, Zhang Y, Zou D. Mediterranean diet adherence and incident acute pancreatitis: a prospective cohort study. Therap Adv Gastroenterol 2025; 18:17562848251346291. [PMID: 40520452 PMCID: PMC12166277 DOI: 10.1177/17562848251346291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 05/14/2025] [Indexed: 06/18/2025] Open
Abstract
Background The relationship between Mediterranean diet (MedDiet) adherence and acute pancreatitis (AP) risk is largely unknown. Objectives To investigate the associations between MedDiet adherence and AP risk and joint associations of genetic risk and MedDiet adherence with AP risk. Design A prospective cohort study using data from UK Biobank, a large population-based prospective study that recruited over 500,000 participants aged 40-69 between 2006 and 2010 across the United Kingdom. Methods We included 103,449 participants free of AP with typical dietary intake from 24-h dietary recalls. MedDiet adherence was measured via the Mediterranean Diet Adherence Screener (MEDAS) continuous score. Genetic predisposition to AP was estimated by polygenic risk score (PRS). Incident AP cases were identified via electronic medical records. Hazard ratios (HRs) with 95% confidence intervals (CIs) were estimated by Cox proportional hazards models. Mediation analyses were further applied to explore the mediating effects of the low-grade inflammation (INFLA) score and metabolic status. Results Over a mean follow-up period of 10.4 years, 371 AP cases were documented. Higher MedDiet adherence defined by MEDAS continuous score was inversely associated with lower AP risk (highest vs lowest tertiles: HR 0.60, 95% CI 0.46-0.79, p < 0.001), with the INFLA score and metabolic status mediating 10% and 7.1% of the association, respectively. Although no interaction was observed between PRS and MedDiet adherence, participants with combined low genetic risk and the highest MedDiet adherence had the lowest risk of AP (HR 0.54, 95% CI 0.36-0.80, p = 0.002). Conclusion The study suggests that higher adherence to the MedDiet is associated with a decreased risk of AP, which is partially mediated by inflammation and metabolic status, and may attenuate the deleterious impact of genetics on AP risk.
Collapse
Affiliation(s)
- Chunhua Zhou
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiawei Geng
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhipeng Wu
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lintao Dan
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hanyi Huang
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xixian Ruan
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jie Chen
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 866, Yuhangtang Road, Changsha 410013, China
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Yao Zhang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Rui Jin Er Road, Shanghai 200025, China
| | - Duowu Zou
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Rui Jin Er Road, Shanghai 200025, China
| |
Collapse
|
12
|
Jeiroshi A, Deng J, Xu Z, Comandatore A, Xu G, Glaviano A, Morelli L, Garajova I, Giovannetti E. Navigating the Paradox of Senescence and Chemoresistance in Pancreatic Cancer. Semin Cancer Biol 2025:S1044-579X(25)00088-4. [PMID: 40513693 DOI: 10.1016/j.semcancer.2025.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/18/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
Cellular senescence, described as a mechanism of irreversible cell cycle arrest, has emerged as a complex and multifaceted process with significant implications in cancer biology, particularly in pancreatic ductal adenocarcinoma (PDAC). This literature review aims to explore the intricate role of senescence in PDAC, focusing on its dual nature during tumorigenesis, in addition to therapy resistance, and its potential as a therapeutic target. Senescence escape was found to play a crucial role in PDAC progression, prompting the development of various pro-senescence therapies. However, recent studies have revealed a paradoxical aspect of the senescence-associated secretory phenotype, revealing its pro-tumorigenic effects and contribution to immune evasion in PDAC. By integrating insights from recent molecular studies, this review synthesizes current knowledge on the role of senescence in PDAC tumorigenesis and chemoresistance, with an emphasis on the emerging role of the tumor microenvironment and explores current and promising avenues for future research and potential therapeutic interventions.
Collapse
Affiliation(s)
- Amal Jeiroshi
- Master Oncology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Juan Deng
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Ziyao Xu
- Senior Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Annalisa Comandatore
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, 56100, Italy
| | - Geng Xu
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Luca Morelli
- Senior Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Ingrid Garajova
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy.
| |
Collapse
|
13
|
Gao Y, Mi J. Causal mediation of plasma metabolomics in pancreatitis: A Mendelian randomization study. Medicine (Baltimore) 2025; 104:e42557. [PMID: 40489851 DOI: 10.1097/md.0000000000042557] [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] [Indexed: 06/11/2025] Open
Abstract
Pancreatitis frequently leads to hospital stay for digestive system disorders and is in high demand for treatment. To identify possible treatment targets, we utilized Mendelian randomization (MR) to investigate the potential causal effects of metabolites on the outcomes of pancreatitis and examined the intermediary roles of risk factors associated with pancreatitis. We gathered GWAS data on 1091 plasma metabolites and 319 metabolite ratios, along with risk factors and phenotypes associated with pancreatitis and its subtypes. Risk factors included H, T2DM, body mass index (BMI), HLP, cholelithiasis, and Inflammatory bowel disease (IBD). Phenotypic outcomes encompassed acute pancreatitis (AP), chronic pancreatitis, alcohol-induced acute pancreatitis, and alcohol-induced chronic pancreatitis. To test the robustness of the findings, we estimated causality using inverse-variance-weighted MR complemented by sensitivity analyses. Additionally, we performed reverse MR analysis to explore potential reverse causality. This study identified 53 plasma metabolites and 22 metabolite ratios predicted by genetics that were significantly associated with pancreatitis (P < .05). Additionally, 54 metabolite ratios and 193 metabolites were associated with pancreatitis risk factors, with 86 and 27 metabolites, respectively, showing significant associations. The MR analysis confirmed that BMI, IBD, and HLP as pancreatitis risk factors (P < .05). It was also revealed that BMI and IBD mediate the relationship between certain metabolite levels and pancreatitis. The identified metabolites and their ratios have the potential to serve as circulating biomarkers with promising applications in CP screening and prevention strategies.
Collapse
Affiliation(s)
- Yuge Gao
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jia Mi
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
14
|
Pawelec N, Durko Ł, Małecka-Wojciesko E. Changes Connected to Early Chronic Pancreatitis and Early Pancreatic Cancer in Endoscopic Ultrasonography (EUS): Clinical Implications. Cancers (Basel) 2025; 17:1891. [PMID: 40507369 PMCID: PMC12153725 DOI: 10.3390/cancers17111891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2025] [Revised: 05/21/2025] [Accepted: 05/30/2025] [Indexed: 06/16/2025] Open
Abstract
Chronic pancreatitis (CP) is a progressive condition that is associated with severe complications. Diagnosis of late CP is easy due to characteristic clinical presentation and pathognomonic imaging findings, such as pancreatic calcifications. Early changes, such as lobularity and a dilated main pancreatic duct, are very subtle and challenging to detect with ultrasonography (US) or even computed tomography (CT). Data were accumulating on the usefulness of EUS in the early diagnosis of CP. The sensitivity values for detecting early CP (ECP) by US, MRI, and EUS were 67-69%, 77-78%, and 81-84%, respectively. The specificity values for detecting ECP by US, MRI, and EUS were 90-98%, 83-96%, and 90-100%, respectively. Pancreatic cancer (PDAC) is one of the leading cancers worldwide, with increasing morbidity. Due to its poor prognosis and survival, early diagnosis is crucial. For this indication, EUS also shows better outcomes compared to other imaging methods, especially in tumors < 2 cm. The sensitivity and specificity for diagnosing PDAC with MRI and EUS were 52.3-93%, 77.1-89%, 72-100%, and 90%, respectively. In addition, EUS can detect precancerous conditions that are associated with a higher risk of PDAC. EUS-assisted new techniques, like elastography and contrast enhancement, facilitate the diagnosis of pancreatic lesions and make it even more accurate. Early PDAC changes, such as main pancreatic duct dilatation or irregular margins of pancreatic solid masses, may be detected with EUS. This review describes the efficacy of different imaging techniques in the early detection of CP and PDAC. In addition, we describe the useful interventions made possible by early diagnosis of PDAC and CP.
Collapse
Affiliation(s)
| | | | - Ewa Małecka-Wojciesko
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-153 Lodz, Poland; (N.P.); (Ł.D.)
| |
Collapse
|
15
|
Boubaddi M, Rossi J, Marichez A, Marty M, Amintas S, Laurent C, Dabernat S. Preoperative Prognostic Factors in Resectable Pancreatic Cancer: State of the Art and Prospects. Ann Surg Oncol 2025; 32:4117-4127. [PMID: 40095311 DOI: 10.1245/s10434-025-17062-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/09/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Only 15% to 20% of patients with pancreatic ductal adenocarcinoma (PDAC) have access to surgical resection, which represents the only chance of curative treatment. Current resection classifications are almost exclusively anatomic and do not correlate sufficiently with patient survival. It is essential to develop preoperative prognostic factors to distinguish patients at high risk of early postoperative recurrence from those who will have prolonged survival after surgery. In some cases, PDACs may present biomolecular differences reflecting their aggressiveness that are not yet assessable by the current clinical-biologic assessment. This study aimed to assess the preoperative prognostic factors that are already available and the future perspectives being developed. METHOD This study reviewed the literature using the PubMed public database for preoperative prognostic factors for resectable PDAC. CONCLUSION Validated preoperative prognostic factors, whether clinical, biologic, radiologic, or histologic, are very important in anticipating the course of each patient's disease. The identification of potential new prognostic biomarkers such as genomic, transcriptomic, and proteomic analyses and the dosage of circulating tumor DNA are very serious avenues to be developed, but the extraction and analysis techniques as well as the interpretation of their results need to be standardized in prospective studies.
Collapse
Affiliation(s)
- Mehdi Boubaddi
- Colorectal Unit, Department of Digestive Surgery, Bordeaux University Hospital, Bordeaux, France.
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, Bordeaux, France.
- Hepatobiliary and Pancreatic Surgery Department, Bordeaux University Hospital, Bordeaux, France.
| | - Julia Rossi
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, Bordeaux, France
| | - Arthur Marichez
- Colorectal Unit, Department of Digestive Surgery, Bordeaux University Hospital, Bordeaux, France
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, Bordeaux, France
| | - Marion Marty
- Tumor Biology and Tumor Bank Laboratory, CHU Bordeaux, Bordeaux, France
| | - Samuel Amintas
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, Bordeaux, France
| | - Christophe Laurent
- Colorectal Unit, Department of Digestive Surgery, Bordeaux University Hospital, Bordeaux, France
| | - Sandrine Dabernat
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, Bordeaux, France
| |
Collapse
|
16
|
Yujin T, Dandan D, Qian Z, Wenhao P, Xingwei D. Epidemiological and demographic drivers of alcohol-attributable pancreatitis from 1990 to 2021: Findings from the 2021 Global Burden of Disease study. Alcohol 2025; 125:67-78. [PMID: 40122354 DOI: 10.1016/j.alcohol.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/11/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Alcohol significantly contributes to pancreatitis, causing high global mortality and health burden. This study examines trends in alcohol-attributable pancreatitis (AAP) from 1990 to 2021 using Global Burden of Disease (GBD) 2021 data, focusing on demographic, temporal, and regional variations to inform policymaking. METHODS AAP-related deaths and disability-adjusted life years (DALYs) were analyzed across 204 countries from 1990 to 2021, stratified by Sociodemographic Index (SDI), gender, and age groups. An age-period-cohort model assessed age-standardized DALY rates (ASDR), and decomposition analysis quantified impacts of population growth, aging, and epidemiological changes. RESULTS AAP-related DALYs rose from 401,700 in 1990 to 699,300 in 2021, though ASDR and ASMR showed declines globally. Burden increased notably in low and lower-middle SDI regions, especially among those under 40, while high SDI regions achieved better control. Males faced a disproportionately high burden due to alcohol consumption patterns, although some regions saw rising female burdens. Low-SDI areas suffered from limited healthcare, increasing alcohol use, and weak policies, with younger populations contributing significantly to rising burdens. Projections estimate 1.146 million DALYs annually by 2050, with males comprising over 90%. A GBD-AAP visualization platform was developed to present burden data and trends. CONCLUSIONS AAP exhibits significant regional and gender disparities. Targeted measures, including alcohol regulation, resource allocation, and public health education, are critical in low-SDI regions and among young males to mitigate AAP burden. The GBD-AAP platform offers valuable tool for targeted interventions.
Collapse
Affiliation(s)
- Tang Yujin
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, FoShan, 528000, China
| | - Dai Dandan
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhong Qian
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Pan Wenhao
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Di Xingwei
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China.
| |
Collapse
|
17
|
Abd El-Fattah AA, Hamid Sadik NA, Shahin AM, Shahin NN. Simvastatin and eugenol restore autophagic flux and alleviate oxidative, inflammatory, and fibrotic perturbations in an arginine-induced chronic pancreatitis rat model. Arch Biochem Biophys 2025; 768:110357. [PMID: 40015469 DOI: 10.1016/j.abb.2025.110357] [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: 10/25/2023] [Revised: 02/15/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Chronic pancreatitis (CP), a progressive inflammatory disease characterized by pancreatic tissue destruction and fibrosis, is considered a challenging health burden due to insufficiencies of current management procedures. Autophagy impairment has emerged as a major triggering event in pancreatitis, raising interest in exploring the potential of targeting autophagy as a possible interventional strategy. This study aimed to evaluate the possible ameliorative effect of two autophagy modulators, simvastatin and eugenol, on CP-related perturbations in an arginine-induced rat model. Repeated l-arginine administration (5 g/kg divided into 2 doses with a 1 h interval, given intraperitoneally every 3rd day for a total of 10 times) provoked CP features, demonstrated by acinar damage, oxidative stress, inflammation, and fibrosis. Arginine-triggered pancreatitis was accompanied by hampered pancreatic autophagic flux, evidenced by overexpression of pancreatic p62 and LC3-Ⅱ and downregulation of pancreatic AMPK and LAMP-1 mRNA expression. Treatment with simvastatin (20 mg/kg, intraperitoneally 24 h, before each arginine dose) and eugenol (50 mg/kg/day orally for 30 days) achieved significant anti-oxidative, anti-inflammatory, and anti-fibrotic effects, and reversed the arginine-instigated autophagic blockade, with superior ameliorative effects attained by eugenol. Altogether, simvastatin and eugenol provide a promising interventional approach for CP, at least partly, by restoring the impaired autophagic flux associated with CP.
Collapse
Affiliation(s)
| | | | - Ahmad Mustafa Shahin
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Nancy Nabil Shahin
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
18
|
Gao X, Zhang G, Wang F, Ruan W, Sun S, Zhang Q, Liu X. Emerging roles of EGFL family members in neoplastic diseases: Molecular mechanisms and targeted therapies. Biochem Pharmacol 2025; 236:116847. [PMID: 40044051 DOI: 10.1016/j.bcp.2025.116847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/09/2025]
Abstract
Epidermal growth factor-like proteins (EGFLs) contain more than a single EGF/EGF-like domain within their protein structure. To date, ten EGFL family members (EGFL1-10) have been characterized across diverse tissues and developmental stages under different conditions. In this review, we conclude that EGFLs are instrumental in regulating biological activities and pathological processes. Under physiological conditions, EGFLs participate in angiogenesis, neurogenesis, osteogenesis, and other processes. Under pathological conditions, EGFLs are linked with different diseases, particularly cancers. Furthermore, we highlight recent advancements in the study of EGFLs in biological conditions and cancers. In addition, the regulatory role and key underlying mechanism of EGFLs in mediating tumorigenesis are discussed. This paper also examines potential antagonists that target EGFL family members in cancer therapeutics. In summary, this comprehensive review elucidates the critical role of EGFLs in neoplastic diseases and highlights their potential as therapeutic targets.
Collapse
Affiliation(s)
- Xiaoge Gao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Guopeng Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Feitong Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Wenhui Ruan
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China
| | - Shishuo Sun
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Qing Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, PR China
| | - Xiangye Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China; Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, PR China.
| |
Collapse
|
19
|
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
|
20
|
Dreyer SB, Beer P, Hingorani SR, Biankin AV. Improving outcomes of patients with pancreatic cancer. Nat Rev Clin Oncol 2025; 22:439-456. [PMID: 40329051 DOI: 10.1038/s41571-025-01019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 05/08/2025]
Abstract
Research studies aimed at improving the outcomes of patients with pancreatic ductal adenocarcinoma (PDAC) have brought about limited progress, and in clinical practice, the optimized use of surgery, chemotherapy and supportive care have led to modest improvements in survival that have probably reached a plateau. As a result, PDAC is expected to be the second leading cause of cancer-related death in Western societies within a decade. The development of therapeutic advances in PDAC has been challenging owing to a lack of actionable molecular targets, a typically immunosuppressive microenvironment, and a disease course characterized by rapid progression and clinical deterioration. Yet, the progress in our understanding of PDAC and identification of novel therapeutic opportunities over the past few years is leading to a strong sense of optimism in the field. In this Perspective, we address the aforementioned challenges, including biological aspects of PDAC that make this malignancy particularly difficult to treat. We explore specific areas with potential for therapeutic advances, including targeting mutant KRAS, novel strategies to harness the antitumour immune response and approaches to early detection, and propose mechanisms to improve clinical trial design and to overcome various community and institutional barriers to progress.
Collapse
Affiliation(s)
- Stephan B Dreyer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- Department of Hepatobiliary Surgery, Royal Liverpool University Hospital, Liverpool, UK
| | - Philip Beer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- Hull York Medical School, University of York, York, UK
| | - Sunil R Hingorani
- Department of Internal Medicine, Division of Hemotology/Oncology, University of Nebraska Medical Center, Omaha, NE, USA
- Pancreatic Cancer Center of Excellence, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK.
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK.
| |
Collapse
|
21
|
Chen JF, Guo SJ, He B, Zheng W, Jiang WJ, Yuan Z, Xiang Y, Peng C, Xiong W, Shi JY. Advances of dual inhibitors based on ALK for the treatment of cancer. Bioorg Chem 2025; 159:108417. [PMID: 40168884 DOI: 10.1016/j.bioorg.2025.108417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 04/03/2025]
Abstract
Anaplastic lymphoma kinase (ALK), which encodes a highly conserved receptor tyrosine kinase (RTK), is important for the development and progression of many tumors, especially non-small cell lung cancer (NSCLC). Currently, third-generation ALK inhibitors are used to treat ALK-mutant NSCLC, but the rapid emergence of resistance during treatment greatly limits their efficacy in clinic. In comparison to single-target inhibitors, ALK dual inhibitors offer the benefits of reducing the emergence of drug resistance, improving treatment efficacy, and optimizing pharmacokinetic features due to the synergistic function of ALK and other associated targets involved in tumor progression. Therefore, we outline the development of ALK dual inhibitors, highlight their design approaches and structure-activity relationship (SAR), and offer insights into new challenges and potential future directions in this area.
Collapse
Affiliation(s)
- Jin-Feng Chen
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731. China; Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Shu-Jin Guo
- Department of Health Management Center, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Bin He
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Wei Zheng
- Department of Integrative Medicine, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Wen-Jie Jiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Zhuo Yuan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu Xiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Cheng Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, School of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Wei Xiong
- Department of urology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu 610072, China.
| | - Jian-You Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| |
Collapse
|
22
|
Xiao Z, Puré E. The fibroinflammatory response in cancer. Nat Rev Cancer 2025; 25:399-425. [PMID: 40097577 DOI: 10.1038/s41568-025-00798-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/06/2025] [Indexed: 03/19/2025]
Abstract
Fibroinflammation refers to the highly integrated fibrogenic and inflammatory responses mediated by the concerted function of fibroblasts and innate immune cells in response to tissue perturbation. This process underlies the desmoplastic remodelling of the tumour microenvironment and thus plays an important role in tumour initiation, growth and metastasis. More specifically, fibroinflammation alters the biochemical and biomechanical signalling in malignant cells to promote their proliferation and survival and further supports an immunosuppressive microenvironment by polarizing the immune status of tumours. Additionally, the presence of fibroinflammation is often associated with therapeutic resistance. As such, there is increasing interest in targeting this process to normalize the tumour microenvironment and thus enhance the treatment of solid tumours. Herein, we review advances made in unravelling the complexity of cancer-associated fibroinflammation that can inform the rational design of therapies targeting this.
Collapse
Affiliation(s)
- Zebin Xiao
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
23
|
Yin H, Zhang Z, Zhang Q, You Y, Zhang Z, Han Y, Zhang Q, You B. PLAU serves as a prognostic biomarker correlated with perineural invasion in HNSCC. Cancer Genet 2025; 294-295:145-155. [PMID: 40319793 DOI: 10.1016/j.cancergen.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 03/25/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
In head and neck squamous cell carcinoma (HNSCC), perineural invasion (PNI) is a distinctive clinicopathologic feature associated with poor survival. To improve patient prognosis, our investigation delved into the underlying mechanism of PNI in HNSCC, especially laryngeal cancer and hypopharyngeal carcinoma. Based on data from the Cancer Genome Atlas (TCGA), genes were categorized into two groups based on the presence or absence of PNI. Plasminogen activator urokinase (PLAU) was screened out as the key molecular. Next, a tissue microarray comprising 68 patients with HNSCC was used to explore the association between PLAU and nerve growth factor (NGF), a positive control of PNI. Then, the co-culture model and cell damage function experiments were used to investigate the carcinogenic effect of PLAU. CCK8 and Transwell assays confirmed the role of PLAU in promoting proliferation and metastasis. The PC12 neurite growth assay and the co-culture system suggested that PLAU influences malignant behaviors by facilitating PNI. Moreover, introducing small molecule compounds to impede PLAU and NGF can effectively revert tumor progression in vivo. PLAU promotes tumor malignancy by facilitating PNI in HNSCC, offering a novel reference for clarifying the molecular mechanisms underlying PNI and identifying potential therapeutic targets for HNSCC.
Collapse
Affiliation(s)
- Haimeng Yin
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China; Medical School of Nantong University, Nantong, 226001, China
| | - Zixiang Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Qing Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Yiwen You
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China; Medical School of Nantong University, Nantong, 226001, China
| | - Zhenxin Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Yumo Han
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China; Medical School of Nantong University, Nantong, 226001, China
| | - Qicheng Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China; Medical School of Nantong University, Nantong, 226001, China.
| | - Bo You
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China; Institute of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China; Medical School of Nantong University, Nantong, 226001, China.
| |
Collapse
|
24
|
Zhang J, Yin R, Xue Y, Qin R, Wang X, Wu S, Zhu J, Li YS, Zhang C, Wei Y. Advances in the study of epithelial mesenchymal transition in cancer progression: Role of miRNAs. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:69-90. [PMID: 40185337 DOI: 10.1016/j.pbiomolbio.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 04/02/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Epithelial-mesenchymal transition (EMT) has been extensively studied for its roles in tumor metastasis, the generation and maintenance of cancer stem cells and treatment resistance. Epithelial mesenchymal plasticity allows cells to switch between various states within the epithelial-mesenchymal spectrum, resulting in a mixed epithelial/mesenchymal phenotypic profile. This plasticity underlies the acquisition of multiple malignant features during cancer progression and poses challenges for EMT in tumors. MicroRNAs (miRNAs) in the microenvironment affect numerous signaling processes through diverse mechanisms, influencing physiological activities. This paper reviews recent advances in EMT, the role of different hybrid states in tumor progression, and the important role of miRNAs in EMT. Furthermore, it explores the relationship between miRNA-based EMT therapies and their implications for clinical practice, discussing how ongoing developments may enhance therapeutic outcomes.
Collapse
Affiliation(s)
- Jia Zhang
- School of Pharmacy, Jiangsu University, Zhen Jiang, 212013, China
| | - Runting Yin
- School of Pharmacy, Jiangsu University, Zhen Jiang, 212013, China.
| | - Yongwang Xue
- School of Pharmacy, Jiangsu University, Zhen Jiang, 212013, China
| | - Rong Qin
- Department of Medical Oncology, Jiangsu University Affiliated People's Hospital, Zhenjiang Clinical Medical College of Nanjing Medical University, Zhenjiang, China
| | - Xuequan Wang
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Shuming Wu
- School of Pharmacy, Jiangsu University, Zhen Jiang, 212013, China
| | - Jun Zhu
- School of Pharmacy, Jiangsu University, Zhen Jiang, 212013, China
| | - Yan-Shuang Li
- Department of Breast Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Cai Zhang
- School of Pharmacy, Jiangsu University, Zhen Jiang, 212013, China
| | - Yuan Wei
- School of Pharmacy, Jiangsu University, Zhen Jiang, 212013, China.
| |
Collapse
|
25
|
Britton E, Kobetic M, McNally E, Rudd S, Potter S, Hinchliffe R, Rees J. A systematic review of clinical outcome reporting for curative surgical treatment of patients with pancreatic adenocarcinoma. HPB (Oxford) 2025; 27:758-770. [PMID: 40122766 DOI: 10.1016/j.hpb.2025.02.008] [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] [Received: 05/01/2024] [Revised: 12/06/2024] [Accepted: 02/15/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Pancreatic cancer is a leading cause of cancer-related death. Surgery (with systemic therapy) provides the only chance for long-term survival, but carries a high risk of morbidity and mortality. Robust evidence from meta-analyses, essential in informing decisions, is thwarted by inconsistencies between studies. This systematic review determines the nature and degree of heterogenous outcome reporting in research evaluating curative pancreatic cancer surgery. METHODS A literature search of Medline, Embase, Cochrane Central and clinicaltrials.gov from 2017 to 2023 for eligible randomised and prospective studies adhering to a PROSPERO registered protocol. RESULTS Included were 156 studies reporting a total of 2088 outcomes which deduplicated to 399 unique endpoints. No single outcome was reported in all studies. 45 % were not defined. Adverse events and delivery of care measures (typically technical aspects of surgery) accounted for 60 % and 32 % of outcomes respectively, compared to 6 % evaluating physical functioning post-surgery. CONCLUSION The vast number and diversity of outcomes in use demonstrates lack of discernment in choice and disparity over domains of importance. Further work is needed to embed uniform outcome definitions, harmonise data collection and refocus research on fewer outcomes of proven relevance. Developing consensus on these critical outcomes through a Core Outcome Set is recommended.
Collapse
Affiliation(s)
- Emily Britton
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol Royal Infirmary, Bristol, BS1 3NU, United Kingdom; University of Bristol Medical School, Learning & Research Centre, Southmead Hospital, Bristol, BS10 5FN, United Kingdom
| | - Matthew Kobetic
- University of Bristol Medical School, Learning & Research Centre, Southmead Hospital, Bristol, BS10 5FN, United Kingdom
| | - Eleanor McNally
- University of Bristol Medical School, Learning & Research Centre, Southmead Hospital, Bristol, BS10 5FN, United Kingdom
| | - Sarah Rudd
- North Bristol NHS Trust, Southmead Hospital, Bristol, BS10 5NB, United Kingdom
| | - Shelley Potter
- University of Bristol Medical School, Learning & Research Centre, Southmead Hospital, Bristol, BS10 5FN, United Kingdom; North Bristol NHS Trust, Southmead Hospital, Bristol, BS10 5NB, United Kingdom
| | - Robert Hinchliffe
- University of Bristol Medical School, Learning & Research Centre, Southmead Hospital, Bristol, BS10 5FN, United Kingdom; North Bristol NHS Trust, Southmead Hospital, Bristol, BS10 5NB, United Kingdom
| | - Jonathan Rees
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol Royal Infirmary, Bristol, BS1 3NU, United Kingdom; University of Bristol Medical School, Learning & Research Centre, Southmead Hospital, Bristol, BS10 5FN, United Kingdom.
| |
Collapse
|
26
|
Fang B, Lu Y, Li X, Wei Y, Ye D, Wei G, Zhu Y. Targeting the tumor microenvironment, a new therapeutic approach for prostate cancer. Prostate Cancer Prostatic Dis 2025; 28:260-269. [PMID: 38565910 DOI: 10.1038/s41391-024-00825-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND A growing number of studies have shown that in addition to adaptive immune cells such as CD8 + T cells and CD4 + T cells, various other cellular components within prostate cancer (PCa) tumor microenvironment (TME), mainly tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs) and myeloid-derived suppressor cells (MDSCs), have been increasingly recognized as important modulators of tumor progression and promising therapeutic targets. OBJECTIVE In this review, we aim to delineate the mechanisms by which TAMs, CAFs and MDSCs interact with PCa cells in the TME, summarize the therapeutic advancements targeting these cells and discuss potential new therapeutic avenues. METHODS We searched PubMed for relevant studies published through December 10 2023 on TAMs, CAFs and MDSCs in PCa. RESULTS TAMs, CAFs and MDSCs play a critical role in the tumorigenesis, progression, and metastasis of PCa. Moreover, they substantially mediate therapeutic resistance against conventional treatments including anti-androgen therapy, chemotherapy, and immunotherapy. Therapeutic interventions targeting these cellular components have demonstrated promising effects in preclinical models and several clinical trials for PCa, when administrated alone, or combined with other anti-cancer therapies. However, the lack of reliable biomarkers for patient selection and incomplete understanding of the mechanisms underlying the interactions between these cellular components and PCa cells hinder their clinical translation and utility. CONCLUSION New therapeutic strategies targeting TAMs, CAFs, and MDSCs in PCa hold promising prospects. Future research endeavors should focus on a more comprehensive exploration of the specific mechanisms by which these cells contribute to PCa, aiming to identify additional drug targets and conduct more clinical trials to validate the safety and efficacy of these treatment strategies.
Collapse
Affiliation(s)
- Bangwei Fang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Ying Lu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaomeng Li
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Gonghong Wei
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| |
Collapse
|
27
|
Syed-Abdul MM, Tian L, Hegele RA, Lewis GF. Futility of plasmapheresis, insulin in normoglycaemic individuals, or heparin in the treatment of hypertriglyceridaemia-induced acute pancreatitis. Lancet Diabetes Endocrinol 2025; 13:528-536. [PMID: 40147461 DOI: 10.1016/s2213-8587(25)00028-2] [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] [Received: 11/25/2024] [Revised: 01/29/2025] [Accepted: 01/29/2025] [Indexed: 03/29/2025]
Abstract
There is a well-established link between the severity of hypertriglyceridaemia and acute pancreatitis and long-term triglyceride-lowering therapies known to prevent episodes of acute pancreatitis. Therefore, it has been assumed, without firm evidence, that rapid lowering of plasma triglycerides would be an effective strategy for reducing the clinical severity of acute pancreatitis and improving health outcomes. Therapies, such as intravenous heparin, intravenous insulin in normoglycaemic individuals (with glucose to prevent hypoglycaemia), and plasmapheresis, continue to be widely used as therapeutic interventions to rapidly reduce serum triglyceride concentration. These therapies are all associated with a risk of adverse reactions, require increased resources, and increase health-care costs. Randomised controlled clinical trials of these therapies have generally shown more rapid reductions in plasma triglycerides than conventional supportive care with the patient made nil by mouth. However, these three therapies alone or in combination, have failed to show effectiveness in improving substantial health benefit outcome measures. While we recognise the theoretical basis for rapidly reducing plasma triglycerides in hypertriglyceridaemia-induced pancreatitis-based on our review of studies using heparin, insulin, plasmapheresis, or a combination of these-these strategies overall do not reduce complications associated with acute pancreatitis or the rapidity of disease resolution. Therefore, we do not advocate the use of triglyceride-lowering therapies at this time, pending more convincing evidence.
Collapse
Affiliation(s)
- Majid M Syed-Abdul
- Departments of Medicine and Physiology and Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
| | - Lili Tian
- Departments of Medicine and Physiology and Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Gary F Lewis
- Departments of Medicine and Physiology and Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
28
|
Gao Y, Chen Q, Wu Z, Yuan L. Regulation of pancreatic β cells by exosomes from different sources. Diabetes Res Clin Pract 2025; 224:112222. [PMID: 40324722 DOI: 10.1016/j.diabres.2025.112222] [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: 02/26/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Diabetes is a chronic metabolic disorder with rising global prevalence, particularly in developed and high-income regions. Central to its pathogenesis is the dysfunction of pancreatic β-cells, alongside impaired glucose and lipid metabolism in peripheral insulin-responsive tissues. Exosomes are nano-sized extracellular vesicles essential for intercellular communication and have emerged as pivotal regulators of metabolic homeostasis. Secreted by virtually all cell types, exosomes encapsulate bioactive cargo that reflects their cellular origin and physiological state, thereby exerting diverse functional effects. Recent evidence highlights the role of exosomes derived from the liver, gut, adipose tissue, skeletal muscle, and mesenchymal stem cells in modulating β-cell proliferation, insulin secretion, and survival. In peripheral tissues exosomes also influence insulin sensitivity by regulating glucose and lipid metabolism, ultimately shaping β-cell responses under hyperglycemic conditions. A more comprehensive understanding of exosome-mediated crosstalk between metabolic organs and pancreatic β-cells could pave the way for the development of exosome-based diagnostic tools and therapeutic strategies aimed at improving early detection, prevention, and treatment of the diabetes.
Collapse
Affiliation(s)
- Yuanyuan Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qi Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhuoying Wu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li Yuan
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
29
|
Fahlbusch T, Antoniou G, Höhn P, Tannapfel A, Uhl W, Belyaev O, Munding J. Use of Histomorphological Features for the Prediction of Grade C Fistula after Whipple Procedure: A Difficult Task. Visc Med 2025; 41:110-120. [PMID: 40519766 PMCID: PMC12165639 DOI: 10.1159/000543436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/04/2025] [Indexed: 06/18/2025] Open
Abstract
Introduction The prediction of occurrence and form of a postoperative pancreatic fistula (POPF) could decrease the risk after pancreatic resections. We developed a score based on histomorphological criteria to predict POPF earlier on. In this study, we test the scoring system to differentiate between patients developing grades B and C POPF. Methods We analysed 92 patients who underwent pancreatic head resection and developed a grade B or C POPF postoperatively. There were no exclusion criteria. Pancreatic resection margins were analysed histologically and pancreatic duct size, fibrosis, fat content, and signs of inflammation were transformed into a scoring system. Results Patients with a grade B fistula (n = 48) were compared to patients with grade C fistula (n = 44). The grade C group was characterized by higher mortality (0 vs. 22, p < 0.001), postpancreatectomy haemorrhage (3 vs. 24, p < 0.001), longer stays in intensive care unit (2.6 vs. 21.1, p < 0.001) and in hospital (29.8 vs. 44.6, p < 0.001). The scoring system was unable to predict grade C fistula. We calculated receiver operator characteristics for all assessed characteristics, which revealed an area under the curve of 0.561. The strongest predictors of POPF grade C were pancreatic fat and soft pancreatic tissue. A combination of only these two items led to a statistically significant difference (p < 0.001) and an AUC of 0.771. Conclusion Establishing a score to differentiate between grade B and grade C POPF is a difficult task. Pancreatic fat seems to be the most relevant histomorphological feature to be used in any scoring system, and it should be combined with other clinical features to enhance its precision and validity.
Collapse
Affiliation(s)
- Tim Fahlbusch
- Department of General and Visceral Surgery, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Georgios Antoniou
- Department of General and Visceral Surgery, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Philipp Höhn
- Department of General and Visceral Surgery, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | | | - Waldemar Uhl
- Department of General and Visceral Surgery, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Orlin Belyaev
- Department of General and Visceral Surgery, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Johanna Munding
- Institute of Pathology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
30
|
Yao B, Chen S, Chen X, Zou L, Fan T, Xiao X. Potential therapeutic targets for ovarian hyperstimulation syndrome revealed by proteome-wide mendelian randomization and colocalization analysis. J Reprod Immunol 2025; 169:104537. [PMID: 40393368 DOI: 10.1016/j.jri.2025.104537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/09/2025] [Accepted: 05/03/2025] [Indexed: 05/22/2025]
Abstract
Ovarian hyperstimulation syndrome (OHSS) is a severe complication associated with assisted reproductive technologies, characterized by metabolic, immune and vascular disorders. Understanding the molecular mechanisms underlying OHSS could reveal potential therapeutic targets and improve patient outcomes. In this study, We aimed to utilize proteome-wide Mendelian randomization (MR) and colocalization analysis to identify plasma proteins associated with OHSS and evaluate their potential as therapeutic targets through druggability assessment. We employed proteome-wide MR analysis summary data-based Mendelian randomization (SMR) analysis and phenome-wide association study (PheWAS) analysis to establish causal relationships between plasma proteins and OHSS. Colocalization analysis confirmed overlaps between proteins and genetic signals associated with OHSS. Pathway and network analyses were conducted to explore biological functions and protein interactions, while drug-target databases were queried for potential therapeutic interventions. Our results showed that 4 key proteins, including Suprabasin (SBSN), SLAMF4 (CD244), Enolase 3 (ENO3) and Thioredoxin domain-containing protein 12 (TXNDC12) were identified as significant contributors to OHSS. Pathway enrichment and interaction analyses further supported their involvement in metabolic, immune and structural pathways related to OHSS. Drug availability for colocalized proteins reveled potential drug targets for ENO3 (2-deoxy-D-glucose), CD244 (lenalidomide) and TXNDC12 (Auranofin), while no potential drug targets were identified for SBSN. Over all, our study identified15 plasma proteins, including SBSN, CD244, ENO3, and TXNDC12, as key contributors to the risk of OHSS through MR and colocalization analysis. These proteins were involved in metabolic regulation, immune response and antioxidant pathways, highlighting potential therapeutic targets and suggesting new directions for treatment strategies.
Collapse
Affiliation(s)
- Bo Yao
- Department of General Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Shanshan Chen
- Department of Reproduction, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215000, China
| | - Xuanyi Chen
- Department of Reproduction, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu 215000, China
| | - Linlin Zou
- Department of General Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Tengyang Fan
- Department of General Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China
| | - Xue Xiao
- Department of General Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, China.
| |
Collapse
|
31
|
Pizzocaro E, Vella R, Frigerio I, Giardino A, Regi P, Scopelliti F, Bannone E, Girelli R, Bignotto L, Dal Mas F, Cobianchi L, Di Renzo L, Butturini G. Personalized nutrition after discharge for pancreatic surgery: A study protocol. Nutr Health 2025; 31:347-356. [PMID: 39569447 DOI: 10.1177/02601060241290281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Background: Pancreatic cancer has an increasing incidence and prevalence trend. Its prognosis is negatively affected by poor/altered nutritional status. Therefore, nutritional interventions play a critical/pivotal role at any stage. Despite being included in the prehabilitation items of Enhanced Recovery After Surgery (ERAS) protocols for pancreatic surgery, nutritional recommendations concerning early post-discharge period are lacking. Aim: To assess the acceptability and feasibility of a personalized and standardized food plan for patients following pancreatic surgery. A secondary aim is to determine if adherence to a personalized food plan is linked to improved postoperative outcomes. Methods: A prospective monocentric cohort study will be performed, including 180 patients undergoing pancreatic surgery. Body composition, nutritional, muscular, and hydro-electrolyte status will be assessed with bio-impedance analysis (BIA). Patients will receive a personalized nutritional plan after discharge for the following 45 days. Summary: Detailed nutritional indications have been formulated with broad consensus for the pre-operative (prehabilitation) and postoperative period. No previous study has specifically formulated or validated a nutritional intervention focused on the early post-discharge period. The results of this study might contribute to paving the way for future research and to broaden nutritional recommendations within the ERAS protocol.
Collapse
Affiliation(s)
- Erica Pizzocaro
- PhD School of Applied Medical-Surgical Sciences, University of Tor Vergata, Rome, Italy
- Department of Hepato-Bilio-Pancreatic Surgery- P. Pederzoli Hospital, Peschiera Del Garda, Verona, Italy
| | - Roberta Vella
- Department of Hepato-Bilio-Pancreatic Surgery- P. Pederzoli Hospital, Peschiera Del Garda, Verona, Italy
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Isabella Frigerio
- Department of Hepato-Bilio-Pancreatic Surgery- P. Pederzoli Hospital, Peschiera Del Garda, Verona, Italy
- Collegium Medicum, University of Social Sciences, Lodz, Poland
| | - Alessandro Giardino
- Department of Hepato-Bilio-Pancreatic Surgery- P. Pederzoli Hospital, Peschiera Del Garda, Verona, Italy
| | - Paolo Regi
- Department of Hepato-Bilio-Pancreatic Surgery- P. Pederzoli Hospital, Peschiera Del Garda, Verona, Italy
| | - Filippo Scopelliti
- Department of Hepato-Bilio-Pancreatic Surgery- P. Pederzoli Hospital, Peschiera Del Garda, Verona, Italy
| | - Elisa Bannone
- Department of Hepato-Bilio-Pancreatic Surgery- P. Pederzoli Hospital, Peschiera Del Garda, Verona, Italy
| | - Roberto Girelli
- Department of Hepato-Bilio-Pancreatic Surgery- P. Pederzoli Hospital, Peschiera Del Garda, Verona, Italy
| | - Laura Bignotto
- Department of Hepato-Bilio-Pancreatic Surgery- P. Pederzoli Hospital, Peschiera Del Garda, Verona, Italy
| | - Francesca Dal Mas
- Collegium Medicum, University of Social Sciences, Lodz, Poland
- Department of Management - Venice School of Management, Ca' Foscari University, Venice, Italy
| | - Lorenzo Cobianchi
- Pancreas Unit Directorship, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia, Italy
| | - Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - Giovanni Butturini
- Department of Hepato-Bilio-Pancreatic Surgery- P. Pederzoli Hospital, Peschiera Del Garda, Verona, Italy
| |
Collapse
|
32
|
Wlaź P, Fitzgerald PJ, Żmudzki P, Socała K. Investigating whether alcohol is transformed to norepinephrine or dopamine in the mouse brain. Pharmacol Rep 2025; 77:703-715. [PMID: 40069538 DOI: 10.1007/s43440-025-00708-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 05/13/2025]
Abstract
BACKGROUND A number of rodent studies have investigated the effects of alcohol (ethanol) administration on the catecholaminergic neurotransmitters, norepinephrine (NE) and dopamine (DA). These studies suggest that presentation of alcohol to mice or rats can alter brain levels of NE and DA, in various subregions. Other studies have presented the hypothesis that there may be an unidentified pathway in rodents, and other organisms, that actually transforms ethanol to NE or DA. Here, this paper investigates the hypothesis in male CD-1 mice. METHODS Experimental mice were systemically injected with an intoxicating dose of stable isotope-labeled carbon 13 (C13) ethanol (ethanol-1-13C, 20% v/v, 1.5 g/kg, ip), and brain samples (hippocampus and brainstem) were collected two hours post-injection. Two other groups of mice received normal unlabeled carbon 12 (C12) ethanol or a water (Control) injection, respectively. RESULTS Although we had difficulty detecting the two neurotransmitters (especially C13 NE) due to their very low concentrations, high-resolution mass spectrometry analysis suggests that C12 ethanol selectively boosted hippocampal C12 NE, and C13 ethanol likewise boosted hippocampal C13 NE. We did not observe effects on DA. CONCLUSIONS These data provide preliminary information on whether there is a novel biosynthetic pathway in mice that converts alcohol to catecholamines in select brain regions, where the ethanol molecule would presumably help form the ethanolamine side chain of NE. There are, however, alternative interpretations of these findings, including that acute alcohol administration modulates catecholamine release, reuptake, metabolism, or canonical biosynthesis.
Collapse
Affiliation(s)
- Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland
| | | | - Paweł Żmudzki
- Department of Medicinal Chemistry, Medical College, Jagiellonian University, Medyczna 9, 30-688, Kraków, Poland
| | - Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland.
| |
Collapse
|
33
|
Wang F, Xu X, Xu J, Li F, Zhang H, Wang L, Yu D. Exploring the value of multiparametric quantitative MRI in the assessment of pancreatic ductal adenocarcinoma fibrosis grading. Eur Radiol 2025; 35:3625-3637. [PMID: 39699670 DOI: 10.1007/s00330-024-11246-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/29/2024] [Accepted: 10/28/2024] [Indexed: 12/20/2024]
Abstract
OBJECTIVES To analyze the performance of multiparametric magnetic resonance imaging (MRI) in quantification of pancreatic ductal adenocarcinoma (PDAC) fibrosis grading. METHOD This prospective study enrolled 79 patients with PDAC confirmed by pathology. Multiparametric MRI including native T1 mapping, intravoxel incoherent motion diffusion-weighted imaging (IVIM-DWI), diffusion kurtosis imaging diffusion-weighted imaging (DKI-DWI), and enhanced T1 mapping were performed before surgery. Masson staining was used to evaluate intratumoral fibrosis content and classified into low- and high-fibrosis groups. MRI parameters were compared between the two groups using multivariable logistic regression analysis. The correlations between fibrosis content and MRI parameters were evaluated using Pearson's correlation. RESULTS D, f, mean diffusion (MD), and enhanced T1 mapping were lower in the high-fibrosis group than in the low-fibrosis group (p < 0.001, p < 0.001, p < 0.001, p = 0.026, respectively). Native T1 mapping and extracellular volume (ECV) were opposite (All p < 0.001). No significant differences in the rest. Multivariable logistic regression revealed that native T1 mapping, MD, and ECV were independent discriminators for PDAC fibrosis grading (p = 0.037, p = 0.031, p = 0.014, respectively); the area under the curve (AUC) of native T1 mapping, MD and ECV was 0.863, 0.798, and 0.929. Among them, ECV had an extremely strong positive correlation with intratumoral fibrosis content. Native T1 mapping and MD were correlated strongly with fibrosis content (positive and negative, respectively). CONCLUSIONS ECV had the highest assessing performance for grading fibrosis in PDAC compared to other MRI parameters, and has the potential to be an imaging biomarker for predicting the fibrosis content of PDAC. KEY POINTS Question The relationship between fibrosis grade of PDAC and quantitative MRI parameters based on T1 mapping and diffusion imaging has not been fully investigated. Findings ECV performed the best in distinguishing between fibrosis grade and increased as interstitial fibrosis increased; clinical indicators offered no added value. Clinical relevance Quantitative MRI parameters provide significant value in evaluating the fibrosis grade of PDAC, which bears significant implications for preoperative risk stratification and the selection of personalized treatment strategies for patients.
Collapse
Affiliation(s)
- Fangqing Wang
- Department of Radiology, Qilu Hospital, Shandong University, Jinan, China
| | - Xinghua Xu
- Department of Radiology, Qilu Hospital, Shandong University, Jinan, China
| | - Jianwei Xu
- Department of Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Feng Li
- Department of Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Hui Zhang
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, China
| | - Lei Wang
- Department of Surgery, Qilu Hospital, Shandong University, Jinan, China.
| | - Dexin Yu
- Department of Radiology, Qilu Hospital, Shandong University, Jinan, China.
| |
Collapse
|
34
|
Sigirli S, Karakas D. Fibrotic Fortresses and Therapeutic Frontiers: Pancreatic Stellate Cells and the Extracellular Matrix in Pancreatic Cancer. Cancer Med 2025; 14:e70788. [PMID: 40437741 PMCID: PMC12119906 DOI: 10.1002/cam4.70788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/19/2025] [Accepted: 03/08/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is characterized by a unique tumor microenvironment (TME) that plays pivotal roles in cancer progression, angiogenesis, metastasis, and drug resistance. This complex and dynamic ecosystem comprises cancer cells, stromal cells, and extracellular matrix (ECM) components, which interact synergistically to drive cancer aggressiveness. Among the stromal cells, cancer-associated fibroblasts (CAFs) and pancreatic stellate cells (PSCs), mainly accepted as a group of CAFs, are central players in shaping the desmoplastic, hypoxic, and immunosuppressive stroma of PDAC. PSCs, the most abundant stromal cells in PDAC, are resident pancreatic cells that undergo phenotypic changes upon activation, driving tumor progression through the secretion of cytokines, growth factors, ECM components (e.g., collagen, hyaluronic acid, fibronectin), and matrix metalloproteinases. In addition to cellular elements, ECM components significantly contribute to cancer aggressiveness by forming a physical barrier that hinders drug penetration, activating signaling pathways through specific receptor interactions, and generating peptides originating from the fragmentation of proteins to induce cancer migration. Regarding their critical roles in tumor progression, therapeutic approaches targeting PSCs and the ECM have garnered increasing interest in recent years. However, PSCs and stromal components may exhibit dual roles, with the potential to both promote and suppress tumor progression under different conditions. Therefore, targeting PSCs or stroma may lead to unintended outcomes, including exacerbation of cancer aggressiveness. METHODS This review focuses on the multifaceted roles of PSCs in PDAC, particularly their interactions with cancer cells and their contributions to therapy resistance. Additionally, we discuss current and emerging therapeutic strategies targeting PSCs and the ECM components, including both preclinical and clinical efforts. CONCLUSION By synthesizing insights from recent literature, this review provides a comprehensive understanding of the role of PSCs in PDAC pathobiology and highlights potential therapeutic approaches targeting PSCs or ECM components to improve patient outcomes.
Collapse
Affiliation(s)
- Sila Sigirli
- Medical Biotechnology, Graduate School of HealthAcibadem Mehmet Ali Aydinlar UniversityIstanbulTurkiye
| | - Didem Karakas
- Medical Biotechnology, Graduate School of HealthAcibadem Mehmet Ali Aydinlar UniversityIstanbulTurkiye
| |
Collapse
|
35
|
Cheng L, Yu H, Qin Y, Wang R, Su T, Lyu G, Huang Z, Li H, Jin Y, Li Y, Wang G. The lncRNA ST18-AS1 suppresses pancreatic cancer progression by enhancing ST18 mRNA stability through anchoring FUS in the cytoplasm. Oncogene 2025:10.1038/s41388-025-03455-4. [PMID: 40450113 DOI: 10.1038/s41388-025-03455-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 05/10/2025] [Accepted: 05/19/2025] [Indexed: 06/03/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with a high mortality rate and short survival time. Long noncoding RNAs (lncRNAs) play a significant role in the progression of PDAC. However, prognostic lncRNAs associated with overall survival (OS) in patients with PDAC remain elusive. RNA sequencing was used to identify differential lncRNA expression between short-term and long-term PDAC patients. We identified a novel lncRNA (ENSG00000253924), termed ST18-AS1 (ST18-associated lncRNA), that is highly expressed in the tissues of long-term PDAC patients. High ST18-AS1 expression was correlated with favorable clinical outcomes, and the upregulation of ST18-AS1 expression in PDAC cell lines suppressed cell proliferation and promoted apoptosis both in vivo and in vitro. The key downstream target regulated by ST18-AS1 was Suppression of tumorigenicity 18 (ST18). We also found that ST18 expression was lower in PDAC tissues compared to non-tumorous adjacent tissues (NATs) and that higher ST18 expression was correlated with better clinical outcomes. Accordingly, the forced expression of ST18 inhibited proliferation and promoted apoptosis in tumor cells. Mechanistic studies showed that ST18-AS1 maintained the stability of ST18 mRNA by binding to Fused in sarcoma (FUS) and anchoring FUS in the cytoplasm. Overall, we identified ST18-AS1 as a novel biomarker that inhibits PDAC cell proliferation and promotes PDAC cell apoptosis through ST18. Targeting ST18-AS1/ST18 may be a potential therapeutic strategy for treating PDAC.
Collapse
Affiliation(s)
- Long Cheng
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hang Yu
- Department of General Surgery, Harbin The First Hospital, Harbin, China
| | - Yujing Qin
- Department of Gastroenterology, Heilongjiang Provincial Hospital, Harbin, China
- Department of Gastroenterology, Heilongjiang Provincial Hospital, Harbin Institute of Technology, Harbin, China
| | - Ruixin Wang
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tie Su
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gaoyuan Lyu
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zijian Huang
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongtao Li
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Jin
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yilong Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Gang Wang
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
36
|
Du R, Wang Y, Pan M, Zhu J, Zhao Y, Zhang C, Liu C, Gao Y. Global, regional, and national burdens of pancreatic cancer attributable to smoking from 1990 to 2021 and the projections to 2035:a systematic analysis from the global burden of disease study 2021. Front Oncol 2025; 15:1547029. [PMID: 40519307 PMCID: PMC12163048 DOI: 10.3389/fonc.2025.1547029] [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: 12/17/2024] [Accepted: 05/05/2025] [Indexed: 06/18/2025] Open
Abstract
Background Studies on global epidemiological patterns regarding the burden of pancreatic cancer (PC) attributable to smoking are limited. This study aimed to analyze the latest disease burden of PC attributable to smoking between 1990 and 2021, further analyze the main factors, and predict the trend in the coming period. Methods Data from the Global Burden of Diseases, Injuries, and Risk Factors Study (GBD) 2021 was extracted and analyzed by different geographical levels, age, sex, and socio-demographic index (SDI). Key measures included age-standardized disability-adjusted life years (DALYs) rates (ASDR), age-standardized mortality rates (ASMR), and average annual percentage changes (AAPCs). Further analyses were conducted using the decomposition analysis and Bayesian Age-Period-Cohort (BAPC) model. Results Globally, the ASDR and ASMR of the disease demonstrated a decreasing trend. The burden was significantly higher among males, the middle-aged, and the elderly population. A positive correlation with SDI levels across most regions was exhibited. Join-point analysis indicated a decreasing trend in disease burden among high SDI regions while an increasing trend among low-middle SDI regions. Decomposition analysis indicated that proactive epidemiological changes had played a positive role in reducing the burden in High SDI regions. Projection analysis estimated that the disease burden in East Asia, showing a significant upward trend, warranted particular focus. Conclusions Despite ongoing tobacco control efforts, the global burden of the disease, which has declined only slightly, remains a significant concern, particularly in high-income areas and among men. Based on stronger tobacco control measures, increased emphasis on disease prevention, early screening, and treatment is essential.
Collapse
Affiliation(s)
- Roubing Du
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Mengshu Pan
- Primary Care Medicine Department, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jie Zhu
- Department of Infectious Disease, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yaping Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Chuanzhen Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Changhong Liu
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yuan Gao
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
37
|
Manoukian P, Damhofer H, Zhao L, van Laarhoven HWM, Bijlsma MF. Stromal Hedgehog Signaling Is Associated with Favorable Outcomes in Pancreatic Cancer. Int J Mol Sci 2025; 26:5200. [PMID: 40508010 PMCID: PMC12154493 DOI: 10.3390/ijms26115200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/20/2025] [Accepted: 05/26/2025] [Indexed: 06/16/2025] Open
Abstract
Aberrant activation of the Hedgehog (Hh) signaling pathway can be observed in various malignancies, particularly in stroma-rich tumors like pancreatic ductal adenocarcinoma (PDAC). In PDAC, Hh signaling is thought to foster an abundant stroma, making it an appealing target for stoma-targeted therapy. However, the use of Hh antagonists in the clinic has thus far not been successful. To reassess the clinical merit of Hh-targeted therapy in PDAC, we sought to better characterize the role of Hh signaling in tumor-stroma crosstalk. Here, we show that Hh ligands are not prognostic per se in PDAC, despite being associated with the favorable classical molecular subtype. Perturbing Hh ligand expression in PDAC cells can effectively alter their trans-signaling capacity but does not impact tumor growth in vivo. However, co-injecting PDAC cells with Smo-proficient MEFs resulted in a significant reduction in xenograft growth, suggesting that Hh-related effects on tumor growth are largely mediated through the stroma. By analyzing transcriptomic sequencing data from co-cultures, comprising human PDAC cells and mouse fibroblasts treated with a Hh-blocking antibody, we could identify stromal hits that are responsive to Hh ligands. We then leveraged the obtained set of genes to allow patient stratification based on stromal response to Hh ligands. We believe that a subset of PDAC patients may benefit from the use of Hh-targeted therapies and thereby encourage the use of our stratification tool to guide their use in PDAC clinical care.
Collapse
Affiliation(s)
- Paul Manoukian
- Laboratory of Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC and University of Amsterdam, 1081 BT Amsterdam, The Netherlands; (P.M.); (H.D.); (L.Z.)
- Cancer Center Amsterdam, Cancer Biology, 1081 BT Amsterdam, The Netherlands;
| | - Helene Damhofer
- Laboratory of Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC and University of Amsterdam, 1081 BT Amsterdam, The Netherlands; (P.M.); (H.D.); (L.Z.)
- Cancer Center Amsterdam, Cancer Biology, 1081 BT Amsterdam, The Netherlands;
- BiOrigin, 2200 Copenhagen, Denmark
| | - Lan Zhao
- Laboratory of Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC and University of Amsterdam, 1081 BT Amsterdam, The Netherlands; (P.M.); (H.D.); (L.Z.)
- Cancer Center Amsterdam, Cancer Biology, 1081 BT Amsterdam, The Netherlands;
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Hanneke W. M. van Laarhoven
- Cancer Center Amsterdam, Cancer Biology, 1081 BT Amsterdam, The Netherlands;
- Department of Medical Oncology, Amsterdam UMC and University of Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Maarten F. Bijlsma
- Laboratory of Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC and University of Amsterdam, 1081 BT Amsterdam, The Netherlands; (P.M.); (H.D.); (L.Z.)
- Cancer Center Amsterdam, Cancer Biology, 1081 BT Amsterdam, The Netherlands;
| |
Collapse
|
38
|
Lee J, Lin T, He Y, Wu Y, Qin J. Toward diffusion MRI in the diagnosis and treatment of pancreatic cancer. Med Oncol 2025; 42:222. [PMID: 40434720 DOI: 10.1007/s12032-025-02759-5] [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: 03/05/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025]
Abstract
Pancreatic cancer is a highly aggressive malignancy with rising incidence and mortality rates, often diagnosed at advanced stages. Conventional imaging methods, such as computed tomography (CT) and magnetic resonance imaging (MRI), struggle to assess tumor characteristics and vascular involvement, which are crucial for treatment planning. This paper explores the potential of diffusion magnetic resonance imaging (dMRI) in enhancing pancreatic cancer diagnosis and treatment. Diffusion-based techniques, such as diffusion-weighted imaging (DWI), diffusion tensor imaging (DTI), intravoxel incoherent motion (IVIM), and diffusion kurtosis imaging (DKI), combined with emerging AI‑powered analysis, provide insights into tissue microstructure, allowing for earlier detection and improved evaluation of tumor cellularity. These methods may help assess prognosis and monitor therapy response by tracking diffusion and perfusion metrics. However, challenges remain, such as standardized protocols and robust data analysis pipelines. Ongoing research, including deep learning applications, aims to improve reliability, and dMRI shows promise in providing functional insights and improving patient outcomes. Further clinical validation is necessary to maximize its benefits.
Collapse
Affiliation(s)
- Junhao Lee
- School of Mathematics and Statistics, Nanjing University of Science and Technology, Nanjing, China
| | - Tingting Lin
- Department of Medical and Radiation Oncology, Affiliated Sanming First Hospital of Fujian Medical University, Sanming, China.
| | - Yifei He
- School of Computer Science and Technology, Nanjing University of Science and Technology, Nanjing, China
| | - Ye Wu
- School of Computer Science and Technology, Nanjing University of Science and Technology, Nanjing, China
| | - Jiaolong Qin
- School of Computer Science and Technology, Nanjing University of Science and Technology, Nanjing, China.
| |
Collapse
|
39
|
Wu Y, Li S, Yu H, Zhang S, Yan L, Guan X, Xu W, Wang Z, Lv A, Tian X, Hao C, Wu J. Integrative Single-Cell and Spatial Transcriptomics Analysis Reveals ECM-remodeling Cancer-associated Fibroblast-Derived POSTN as a Key Mediator in Pancreatic Ductal Adenocarcinoma Progression. Int J Biol Sci 2025; 21:3573-3596. [PMID: 40520021 PMCID: PMC12160862 DOI: 10.7150/ijbs.108618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/10/2025] [Indexed: 06/18/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents significant clinical challenges owing to its dense stroma and complex tumor microenvironment (TME). In this study, large-scale single-cell transcriptomics and spatial transcriptomics (ST) were integrated to dissect the heterogeneity of fibroblasts and their crosstalk with epithelial cells, with a focus on key ligand-receptor interactions. Eight distinct fibroblast subpopulations were identified, among which extracellular matrix (ECM)-remodeling fibroblasts were particularly enriched in tumor tissues and associated with poor prognosis. ECM-remodeling fibroblasts were located at the terminal stage of the fibroblast pseudotime trajectory, and SOX11 was identified as a key transcription factor in this subpopulation. Further analyses revealed that ECM-remodeling fibroblasts can interact with epithelial cells through the POSTN-ITGAV/ITGB5 ligand-receptor axis, a critical pathway that promotes tumor progression. Clinical analyses demonstrated a strong correlation between POSTN expression and poor prognosis in patients with PDAC. Mechanistically, POSTN interacts with integrin ITGAV/ITGB5 on tumor cells, activating the PI3K/AKT/β-catenin pathway and promoting epithelial-mesenchymal transition (EMT) phenotype. Pharmacological inhibition of the POSTN-integrin axis partially reversed these malignant traits, highlighting its potential as a therapeutic target. This study provides new insights into fibroblast heterogeneity and its role in PDAC progression, emphasizing the POSTN-ITGAV/ITGB5 axis as a promising target for therapeutic interventions.
Collapse
Affiliation(s)
- Yifan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shuquan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hao Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Sha Zhang
- Department of Critical Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Liang Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaoya Guan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wei Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhen Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ang Lv
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiuyun Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chunyi Hao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jianhui Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
40
|
Lu Y, Wu L, Yue X, Peng T, Yang M, Chen J, Han P. Quantitative Evaluation of Acute Pancreatitis Based on Dual-Energy Computed Tomography. J Comput Assist Tomogr 2025:00004728-990000000-00460. [PMID: 40423618 DOI: 10.1097/rct.0000000000001768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/21/2025] [Indexed: 05/28/2025]
Abstract
OBJECTIVE To evaluate the value of dual-energy computed tomography (DECT) parameters for the quantitative diagnosis of acute pancreatitis (AP) and classification of its severity. METHODS Patients with AP underwent a plain CT scan and three contrast-enhanced DECT scans. We analyzed the group differences in iodine concentration (IC) and slope of the spectral Hounsfield unit curve (λHU) of the 3-phase enhanced scans (arterial, venous, and delayed phases). RESULTS The study included 60 AP patients (38 males and 22 females; mean age: 47.43±13.47 y). On the basis of the CT severity index (CTSI), the patients were divided into 2 groups: group A (mild AP, n=26) and group B (moderate/severe AP, n=34). IC and λHU in the arterial and venous phases were all significantly higher in group A than in group B (P<0.001) and could effectively differentiate the 2 groups. The areas under the curve were 0.753 (95% CI: 0.624-0.855), 0.799 (95% CI: 0.676-0.892), 0.774 (95% CI: 0.647-0.872), and 0.842 (95% CI: 0.724-0.923) for IC at arterial and venous phases and λHU at arterial and venous phases, respectively. These parameters decreased with the increase of CTSI, showing significant negative correlations, with r were -0.512 (95% CI: -0.678 to -0.297), -0.492 (95% CI: -0.663 to -0.272), -0.552 (95% CI: -0.707 to -0.346), -0.569 (95% CI: -0.719 to -0.368) for IC at arterial and venous phases and λHU at arterial and venous phases, respectively (P<0.001). CONCLUSIONS DECT imaging can quantitatively analyze AP, and the IC and λHU can be used to distinguish mild and severe cases, adding functional information to the CT morphology to determine the severity and prognosis of the disease.
Collapse
Affiliation(s)
- Yuting Lu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging
| | - Linxia Wu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging
| | - Xiaofei Yue
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging
| | - Tao Peng
- Departments of Pancreatic Surgery
| | - Ming Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging
| | - Jinhuang Chen
- Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ping Han
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging
| |
Collapse
|
41
|
Sharma S, Singh R, Kant S, Mishra MK. Integrating AI/ML and multi-omics approaches to investigate the role of TNFRSF10A/TRAILR1 and its potential targets in pancreatic cancer. Comput Biol Med 2025; 193:110432. [PMID: 40424767 DOI: 10.1016/j.compbiomed.2025.110432] [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: 04/15/2025] [Revised: 05/19/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with a five-year survival of under 10 % despite current therapies. Aggressive tumor biology, a desmoplastic stroma that limits drug delivery and immune cell infiltration, and profound resistance to apoptosis make it more complex to treat. Here, we describe a multi-layered system biology and drug discovery pipeline that integrates bulk genomics, single-cell spatial transcriptomics, proteomics, competing endogenous RNA (ceRNA) network analysis, and deep learning-driven quantitative structure-activity relationship (QSAR) modeling. By implementing this pipeline, we predicted that TNFRSF10A encodes for the TRAILR1 death receptor as a potential therapeutic target in PDAC. Mutational and expressional analysis also confirmed TNFRSF10A as a putative target in PDAC. Cancer cells within the PDAC microenvironment exhibit aberrantly elevated TNFRSF10A expression. Immune-excluded tumor niches and pro-survival signaling link this elevated expression. Using an advanced transformer-based deep learning approach, SELFormer, combined with QSAR analysis-based virtual screening, we identified previously unexplored FDA-approved drugs and natural compounds, i.e., Temsirolimus, Ergotamine, and capivasertib, with potential TRAILR1 modulatory effects. During molecular dynamics simulations, these repurposed candidates showed the highest binding affinities against TNFRSF10A for 300 ns. These showed favorable binding energies (MM-PBSA), minimal RMSD drift, PCA, and SASA. We propose TNFRSF10A as a therapeutically important PDAC vulnerability nurtured by spatially resolved expression patterns and dynamic molecular modeling. This study has used a novel integration of AI-implemented chemical modeling, high-throughput screening, and a multi-omics approach to unravel and pharmacologically target a cancer compartment-specific weakness in a notoriously drug-resistant cancer.
Collapse
Affiliation(s)
- Sudhanshu Sharma
- Cancer Research Center, Department of Biological Sciences, Alabama State University, Montgomery, AL, 36104, USA
| | - Rajesh Singh
- Microbiology, Biochemistry, and Immunology, Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Shiva Kant
- Department of Biology and Environmental Sciences, College of Sciences, Auburn University of Montgomery, Montgomery, USA
| | - Manoj K Mishra
- Cancer Research Center, Department of Biological Sciences, Alabama State University, Montgomery, AL, 36104, USA.
| |
Collapse
|
42
|
Thakur R, Mullen NJ, Mehla K, Singh PK. Tumor-stromal metabolic crosstalk in pancreatic cancer. Trends Cell Biol 2025:S0962-8924(25)00109-6. [PMID: 40425415 DOI: 10.1016/j.tcb.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with a dire prognosis. Standard-of-care chemotherapy regimens offer marginal survival benefit and carry risk of severe toxicity, while immunotherapy approaches have uniformly failed in clinical trials. Extensive desmoplasia in the PDAC tumor microenvironment (TME) disrupts blood flow to and from the tumor, thereby creating a nutrient-depleted, hypoxic, and acidic milieu that suppresses the function of antitumor immune cells and imparts chemotherapy resistance. Additionally, recent seminal studies have demonstrated crucial roles for metabolic crosstalk - the exchange of metabolites between PDAC cells and stromal cell populations in the TME - in establishing and maintaining core malignant behaviors of PDAC: tumor growth, metastasis, immune evasion, and therapy resistance. In this review, we provide a conceptual overview of metabolic crosstalk and how it evolves under various selection pressures in the TME, analyze the landscape of proposed tumorigenic metabolic crosstalk pathways, and highlight potentially druggable nodes.
Collapse
Affiliation(s)
- Ravi Thakur
- Department of Oncology Science, University of Oklahoma College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Nicholas J Mullen
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kamiya Mehla
- Department of Oncology Science, University of Oklahoma College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Pankaj K Singh
- Department of Oncology Science, University of Oklahoma College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
43
|
Wang W, Hu K, Xue J, Chen J, Du X, Zhao T, Chen Y, Tang X, Xu L, Hao X, Li X, Yang Y. In vivo FAP-CAR macrophages enhance chemotherapy and immunotherapy against pancreatic cancer by removing the fibrosis barrier. J Control Release 2025:113888. [PMID: 40425095 DOI: 10.1016/j.jconrel.2025.113888] [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: 05/07/2025] [Accepted: 05/24/2025] [Indexed: 05/29/2025]
Abstract
Patients with pancreatic ductal adenocarcinoma (PDAC) derive limited benefits from chemotherapy or immunotherapy, with a five-year survival rate still below 10 %. The key therapeutic challenge is the dense fibrosis barrier driven by activated cancer-associated fibroblasts (CAFs) and their secreted collagen, which impedes drug penetration and characterizes PDAC as an immune-desert tumor. To address this challenge, we developed in vivo chimeric antigen receptor macrophages (FAP-CAR-M) targeting fibroblast activation protein-α (FAP), the marker of activated CAFs, to enhance chemo and immunotherapy against PDAC by removing the fibrosis barrier using mannose-modified mRNA-LNP (MLNP). Our results demonstrate that mRNA-MLNP can efficiently reprogram M2 macrophages into FAP-CAR-M. With the FAP-CAR-M treatment, the activated CAF markers (FAP), collagen volume fraction (CVF), and the type I collagen (col1a1) secretion were decreased by 3-fold, 5-fold, and 4-fold in orthotopic PDAC, respectively. By removing the fibrosis barrier, FAP-CAR-M enhanced the penetration of gemcitabine (GEM) and immune cells, improved PDAC sensitivity to chemo and immunotherapy, and significantly prolonged survival. Therefore, in vivo FAP-CAR-M may represent a potential therapeutic approach to enhance chemo and immunotherapy against PDAC by removing the fibrosis barrier.
Collapse
Affiliation(s)
- Wenguang Wang
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China.
| | - Kaiyuan Hu
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Junjie Xue
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Jingyi Chen
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Xiuli Du
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Tian Zhao
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yiwei Chen
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Xinying Tang
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Lu Xu
- Department of Hematology, The First Affiliated Hospital of Hainan Medical University, No. 31 Longhua Road, Longhua District, Haikou 570102, Hainan Province, China
| | - Xinbao Hao
- Department of Hematology, The First Affiliated Hospital of Hainan Medical University, No. 31 Longhua Road, Longhua District, Haikou 570102, Hainan Province, China
| | - Xianjing Li
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yong Yang
- School of Translational Research, China Pharmaceutical University, Nanjing 211198, China; Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
44
|
Ma J, Xie Y, Teng Z, Jiang L, Liu G. Engineered cell membrane-based nano therapies fight infectious diseases. J Control Release 2025; 384:113884. [PMID: 40418989 DOI: 10.1016/j.jconrel.2025.113884] [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: 04/02/2025] [Revised: 05/18/2025] [Accepted: 05/22/2025] [Indexed: 05/28/2025]
Abstract
Infectious diseases continue to present significant global public health challenges, with pathogens such as bacteria and viruses posing substantial threats to human health. Conventional therapeutic approaches face several limitations, including the rising prevalence of drug resistance, suboptimal targeting, and adverse side effects, which collectively complicate clinical management. Cell membrane vesicles (MVs), characterized by their natural biocompatibility and outstanding drug delivery capabilities, have emerged as a promising platform for addressing these challenges in the treatment of infectious diseases. To further augment the therapeutic potential of MVs, engineering modifications have been extensively employed to enhance their functionality and efficacy. This review provides a comprehensive overview of the production and modification techniques associated with MVs, emphasizing recent advancements in the development of engineered membrane vesicles (EMVs) as versatile nanoplatforms for combating infectious diseases. Additionally, the clinical prospects and existing challenges of EMVs are critically analyzed, and recommendations are proposed to guide future research and facilitate their clinical translation into practical applications in combating infectious disease.
Collapse
Affiliation(s)
- Jiaxin Ma
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yijia Xie
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zihao Teng
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Lili Jiang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
45
|
Romero-Pérez I, Montero JC, Redondo-Puente M, Del Carmen Gómez-García M, Morell-Ginestà M, Capellá G, Pandiella A. An antibody-drug conjugate targeting soluble and membrane-bound TGFα is effective against pancreatic tumors. J Exp Clin Cancer Res 2025; 44:158. [PMID: 40410803 PMCID: PMC12100920 DOI: 10.1186/s13046-025-03421-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 05/15/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Pancreatic cancer is one of the most difficult to treat neoplasias. Because of that, the prognosis of the disease is dismal, and identification of novel therapeutic approaches is needed. This study investigates the role of transforming growth factor-alpha (TGFα) in pancreatic cancer and its potential as a therapeutic target. METHODS Using in silico platforms, it was confirmed that TGFA, the gene encoding TGFα, is significantly overexpressed in pancreatic adenocarcinomas relative to normal pancreatic tissues. In patient-derived xenografts as well as in pancreatic cancer cell lines, multiple molecular forms of TGFα were identified, including the transmembrane TGFα precursor (proTGFα) and the soluble 6 kDa mature form. Functional assays using RNA interference and CRISPR/Cas9 demonstrated that TGFA knockdown significantly impaired cell proliferation, reinforcing the critical role of TGFα in driving tumor growth. The therapeutic potential of targeting TGFα was evaluated through the development of two monoclonal antibodies (5F1 and 16B10) specific for TGFα. RESULTS These antibodies effectively bound to proTGFα-expressing cells, with minimal off-target effects in TGFA-knockout cell lines. When conjugated to cytotoxic agents such as MMAF, the resulting antibody-drug conjugates (ADCs) exhibited potent antiproliferative activity, significantly reducing the viability of TGFα-expressing pancreatic cancer cells. Mechanistic studies revealed that MMAF-loaded ADCs induced G2/M cell cycle arrest, with markers of mitotic disruption evident in treated cells. In vivo, the TGFα-targeting ADCs elicited substantial tumor regression in murine models of pancreatic cancer, whereas the unconjugated antibodies merely stabilized tumor growth. CONCLUSIONS These findings highlight TGFα as a promising therapeutic target in pancreatic cancer, supporting further preclinical and clinical development of TGFα-directed ADCs.
Collapse
Affiliation(s)
- Inés Romero-Pérez
- Instituto de Biología Molecular y Celular del Cáncer- CSIC and CIBERONC, Campus Miguel de Unamuno, Salamanca, 37007, Spain
| | - Juan Carlos Montero
- Instituto de Biología Molecular y Celular del Cáncer- CSIC and CIBERONC, Campus Miguel de Unamuno, Salamanca, 37007, Spain
- Department of Pathologic Anatomy and IBSAL, Salamanca, Spain
| | - Mónica Redondo-Puente
- Instituto de Biología Molecular y Celular del Cáncer- CSIC and CIBERONC, Campus Miguel de Unamuno, Salamanca, 37007, Spain
| | - María Del Carmen Gómez-García
- Instituto de Biología Molecular y Celular del Cáncer- CSIC and CIBERONC, Campus Miguel de Unamuno, Salamanca, 37007, Spain
| | - Mireia Morell-Ginestà
- Hereditary Cancer Program, Catalan Institute of Oncology, Institut d'Investigació Biomèdica de Bellvitge- IDIBELL-ONCOBELL, L'Hospitalet de Llobregat, Barcelona, 08908, Spain
| | - Gabriel Capellá
- Hereditary Cancer Program, Catalan Institute of Oncology, Institut d'Investigació Biomèdica de Bellvitge- IDIBELL-ONCOBELL, L'Hospitalet de Llobregat, Barcelona, 08908, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer- CSIC and CIBERONC, Campus Miguel de Unamuno, Salamanca, 37007, Spain.
| |
Collapse
|
46
|
Zhang BD, Chen X, Su JY, Zhuo SH, Zhao L, Wu JJ, Li WH, Wang TY, Liu L, Yang T, Yang LJ, Zhao YF, Li YM. Rationally designed anti-autophagy nanosystems for reversing the immunosuppressive network in the tumor environment. Nanomedicine (Lond) 2025:1-12. [PMID: 40401367 DOI: 10.1080/17435889.2025.2508133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 05/15/2025] [Indexed: 05/23/2025] Open
Abstract
AIMS To develop a nano-immunotherapy system combining autophagy inhibition and innate immune activation to reverse the immunosuppressive tumor microenvironment (TME) in pancreatic ductal adenocarcinoma (PDAC). MATERIALS & METHODS The pH-responsive polymer PC7A was utilized to co-deliver the autophagy inhibitor chloroquine (CQ) and the STING agonist cyclic diguanylate (CDG), forming the CQCP nanosystem. In vitro and in vivo experiments evaluated autophagy inhibition, MHC-I expression, dendritic cell activation, tumor infiltration of lymphocytes, and survival in PDAC-bearing mice. RESULTS CQCP enhanced MHC-I expression on PDAC cells by 2.1-fold (p < 0.001) and increased activated dendritic cells (CD86+/CD40+) by 3.5-fold (p < 0.01) in the TME. Tumor-infiltrating CD8+ T cells rose by 42.6% (p < 0.001), and systemic immune activation in peripheral lymphoid tissues was observed. CQCP achieved an 86% survival rate in tumor-bearing mice, significantly outperforming monotherapies or free drug combinations. CONCLUSIONS The CQCP system synergistically reverses PDAC immunosuppression by restoring antigen presentation and activating innate immunity. This dual-targeted strategy demonstrates robust antitumor efficacy and offers a promising immunotherapy approach for PDAC.
Collapse
Affiliation(s)
- Bo-Dou Zhang
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Xi Chen
- Zhili College, Tsinghua University, Beijing, China
| | - Jing-Yun Su
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Shao-Hua Zhuo
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Lang Zhao
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Jun-Jun Wu
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Wen-Hao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Tian-Yang Wang
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Ling Liu
- Peking University First Hospital, Beijing, China
| | - Tao Yang
- Key Laboratory of Digestive Disease & Organ Transplantation in Shanxi Province, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Higher Education Key Laboratory of Tumor Immunology & Targeted Drug Development in Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Li-Jun Yang
- Key Laboratory of Digestive Disease & Organ Transplantation in Shanxi Province, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yu-Fen Zhao
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, China
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
- Beijing Institute for Brain Disorders, Beijing, China
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| |
Collapse
|
47
|
Wang T, Song W, Tang Y, Yi J, Pan H. Breaking the immune desert: Strategies for overcoming the immunological challenges of pancreatic cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189353. [PMID: 40412630 DOI: 10.1016/j.bbcan.2025.189353] [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: 11/08/2024] [Revised: 05/18/2025] [Accepted: 05/18/2025] [Indexed: 05/27/2025]
Abstract
Pancreatic cancer is characterised by its highly aggressive nature and extremely poor prognosis, with a uniquely complex tumour immune microenvironment that manifests as a prototypical "immune desert." This immune-desert phenotype primarily arises from the inherently low immunogenicity of the tumour, the formation of a dense fibrotic stroma, severe deficiency in immune cell infiltration, and profound immunosuppressive effects of the metabolic landscape. Specifically, dysregulated tryptophan metabolism, such as indoleamine 2,3-dioxygenase (IDO)-mediated catabolism, and excessive lactate accumulation contribute to impaired T-cell functionality. Collectively, these factors severely limit the efficacy of current immunotherapy strategies, particularly those based on immune checkpoint inhibitors, which have demonstrated significantly lower clinical response rates in pancreatic cancer than in other malignancies. In response to these therapeutic challenges, this review explores integrated treatment strategies that combine metabolic reprogramming, tumour microenvironment remodelling, and next-generation immune checkpoint blockades, such as LAG-3, TIM-3, and VISTA. These emerging approaches hold substantial promise for clinical application. For example, targeting key metabolic pathways, including glycolysis (Warburg effect) and glutamine metabolism, may help restore T-cell activity by alleviating metabolic stress within the tumour milieu. Additionally, localised administration of immune stimulators such as interleukin-12 (IL-12) and CD40 agonists may enhance immune cell infiltration and promote tumour-specific immune activation. Future research should prioritise large-scale, multicentre clinical trials to validate the therapeutic efficacy of these innovative strategies, aiming to achieve meaningful breakthroughs in pancreatic cancer immunotherapy and significantly improve long-term survival and clinical outcomes in affected patients.
Collapse
Affiliation(s)
- Tianming Wang
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China; Zhengning County Community Health Service Center, Qingyang 745300, Gansu Province, China
| | - Wenjing Song
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Yuan Tang
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Jianfeng Yi
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China.
| | - Haibang Pan
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China.
| |
Collapse
|
48
|
Dubrova A, Cavaniol C, Van de Walle A, Mathieu P, Fusilier Z, Yaacoub N, Lalatonne Y, Descroix S, Wilhelm C. Magnetite Nanoparticle Photothermal Therapy in a Pancreatic Tumor-on-Chip: A Dual-Action Approach Targeting Cancer Cells and their Microenvironment. ACS NANO 2025. [PMID: 40397413 DOI: 10.1021/acsnano.5c02099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
The application of magnetite nanoparticles (MagNPs) for photothermal therapy (MagNP-PTT) has recently expanded to cancer treatment. This study introduces MagNP-PTT in a tumor-on-a-chip model to target highly aggressive pancreatic ductal adenocarcinoma (PDAC). A tumor-on-chip system was developed using PANC-1 PDAC cells embedded in a collagen type I extracellular matrix and cultured for 1 week to form tumor spheroids. This platform offers a framework for applying PTT in a model system that aims to mimic the native tumor microenvironment. MagNPs efficiently penetrate the tumor spheroids, achieving controlled heating via near-infrared (NIR) light. By adjusting nanoparticle concentration and laser power, temperature increments of 2 °C between 38-48 °C were established. Temperatures above 44 °C significantly increased cell death, while lower temperatures allowed partial recovery. Beyond inducing cancer cell death, MagNP-PTT altered the extracellular matrix and triggered a slight epithelial-mesenchymal transition marked by increased vimentin expression. These findings highlight MagNP-PTT as a dual-action therapy, targeting both tumor cells and their microenvironment, offering an alternative approach for overcoming stromal barriers in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Anastasiia Dubrova
- Laboratoire Physique des Cellules et Cancer, PCC, CNRS UMR168, Institut Curie, Sorbonne Université, PSL Research University, 75005 Paris, France
| | - Charles Cavaniol
- Laboratoire Physique des Cellules et Cancer, PCC, CNRS UMR168, Institut Curie, Sorbonne Université, PSL Research University, 75005 Paris, France
| | - Aurore Van de Walle
- Laboratoire Physique des Cellules et Cancer, PCC, CNRS UMR168, Institut Curie, Sorbonne Université, PSL Research University, 75005 Paris, France
| | - Paul Mathieu
- Université Sorbonne Paris Nord, Université Paris Cité, Laboratory for Vascular Translational Science, LVTS, INSERM, UMR 1148, Bobigny F-93017, France
| | - Zoé Fusilier
- Institut Curie, PSL University, INSERM U932, Immunity and Cancer, 75005 Paris, France
| | - Nader Yaacoub
- Institut des Molécules et Materiaux du Mans, CNRS UMR-6283, Le Mans Université, F-72085 Le Mans, France
| | - Yoann Lalatonne
- Université Sorbonne Paris Nord, Université Paris Cité, Laboratory for Vascular Translational Science, LVTS, INSERM, UMR 1148, Bobigny F-93017, France
- Département de Biophysique et de Médecine Nucléaire, Assistance Publique-Hôpitaux de Paris, Hôpital Avicenne F- 93009, Bobigny, France
| | - Stephanie Descroix
- Laboratoire Physique des Cellules et Cancer, PCC, CNRS UMR168, Institut Curie, Sorbonne Université, PSL Research University, 75005 Paris, France
| | - Claire Wilhelm
- Laboratoire Physique des Cellules et Cancer, PCC, CNRS UMR168, Institut Curie, Sorbonne Université, PSL Research University, 75005 Paris, France
| |
Collapse
|
49
|
Liu Y, Ran X, Zhou G, Liu Y, Tan W. Multivalent Aptamer Assembly Enhances Tumor-Specific Degradation of Transforming Growth Factor-Beta to Remodel the Stromal and Immunosuppressive Cancer Microenvironment. ACS NANO 2025; 19:18164-18175. [PMID: 40326636 DOI: 10.1021/acsnano.4c16628] [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: 05/07/2025]
Abstract
Extracellular proteins like transforming growth factor-β (TGFβ) are crucial enforcers in the development of cancer stroma and the tumor immunosuppressive microenvironment. Lysosome-targeting chimera-mediated protein degradation appeared as a promising tool for extracellular signal interference but was limited by several lysosome-trafficking receptors and inadequate in vivo degradation efficiency. Here, we designed a multivalent aptamer assembly with a universal pattern to drag extracellular proteins (e.g., TGFβ1) for lysosome degradation with high tumor specificity. By accelerating cell recognition-internalization and lysosomal delivery, the assembly promoted TGFβ blockade and degradation in pancreatic cancer cells and pancreatic stellate cells (PSCs). In vivo, the assembly exhibited highly tumor-specific accumulation and prolonged retention, which resulted in efficient TGFβ inhibition, stromal remodeling, and reversed polarization of immunosuppressive cells in the tumor microenvironment, as well as synergic therapeutic effects when combined with gemcitabine or ovalbumin. Therefore, this study provides a feasible strategy to construct a multivalent aptamer assembly for tumor-specific extracellular protein degradation, after remodeling the tumor stromal and immunosuppressive microenvironment in a manner that enhances the effects of cancer chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Yan Liu
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xinyue Ran
- Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Guangdong Zhou
- Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
50
|
Wang L, Jia Y, Wang J, Xia XH, Wang C. Multiplexed Detection of Pancreatic-Specific Nucleic Acids and Protein Biomarkers Using a Logic Nanofluidic Platform. Anal Chem 2025. [PMID: 40387859 DOI: 10.1021/acs.analchem.5c01978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Early detection of pancreatic cancer is vital for patient survival. However, current diagnostic approaches remain constrained by insufficient precision and specificity inherent to single-biomarker detection strategies. Herein, we develop a nanochannel biosensing platform implementing cooperative dual-signal detection of pancreatic-specific biomarkers CA19-9 and miRNA-196a. Using liquid-liquid interface self-assembly, we constructed anodic aluminum oxide (AAO)-Au hybrid nanochannels integrated with a surface-modified double-key DNA nanolock (DDN). The conformational switch of DDN logic gating triggered by miRNA-196a exposes the CA19-9-aptamer, enabling specific target recognition and consequent ion current signal attenuation. Simultaneously, released miRNA-196a is quantified by catalytic hairpin assembly and hybridization chain reaction-mediated cascade amplification. Experiments show that the present DDN-based logic nanofluidic platform could achieve an ultralow detection limit of 0.000027 U·mL-1 for CA19-9 and 4.74 aM for miRNA-196a, which is 2-3 orders of magnitude higher than traditional ELISA/qPCR methods. Finally, clinical sample analysis confirms the high specificity of this platform in distinguishing pancreatic cancer and acute pancreatitis from healthy individuals. This DDN-functionalized nanofluidic biosensor provides valuable insights into designing precision detection platforms for pancreatic cancer, highlighting its significant potential for clinical diagnostics.
Collapse
Affiliation(s)
- Lina Wang
- State Key Laboratory of Microbial Technology, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yixin Jia
- State Key Laboratory of Microbial Technology, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Jin Wang
- State Key Laboratory of Microbial Technology, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Xing-Hua Xia
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Chen Wang
- State Key Laboratory of Microbial Technology, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|