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 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
|
Blanke EN, Holmes GM. Dysfunction of pancreatic exocrine secretion after experimental spinal cord injury. Exp Neurol 2025; 389:115257. [PMID: 40221007 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] [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
|
3
|
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
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
Dharmasivam M, Zhang S, Zhao X, Richardson V, Wijesinghe TP, Suleymanoglu M, Gholam Azad M, Bernhardt PV, Kaya B, Richardson DR. Advantages of Novel Anti-cancer Selenosemicarbazones: Preferential Reactivity of Their Fe(III), Cu(II), and Zn(II) Complexes with Key Physiological Reductants/Ligands Versus Isosteric Thiosemicarbazones. J Med Chem 2025; 68:9594-9622. [PMID: 40265585 DOI: 10.1021/acs.jmedchem.5c00374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Fe(III) complexes of clinically trialed thiosemicarbazones demonstrate deleterious oxy-myoglobin and oxy-hemoglobin oxidation. Therefore, the PPP4pSe selenosemicarbazone analogues were designed with several PPP4pSe Fe(III) complexes completely preventing deleterious oxy-myoglobin oxidation. This was ascribed to the decreased potentials of their Fe(III) complexes and steric hindrance effects. The Fe(III), Cu(II), and Zn(II) complexes of PPP4pSe demonstrated greater reactivity with physiological reductants/ligands (glutathione, l-cysteine, or l-ascorbate), than respective complexes of the isosteric thiosemicarbazone, PPP4pT. Considering this: (1) [Fe(PPP4pSe)2]+ demonstrated increased reduction relative to [Fe(PPP4pT)2]+ with glutathione and l-cysteine, while l-ascorbate led to comparable reduction; (2) glutathione led to complete dissociation of [Zn(PPP4pSe)2], while incomplete dissociation of [Zn(PPP4pT)2] occurred; and (3) [Cu(PPP4pSe)Cl] demonstrated complete coordinate sphere substitution with glutathione, l-cysteine, and l-ascorbate, whereas [Cu(PPP4pT)Cl] demonstrated partial substitution. The role of glutathione in all three latter reactions is significant, given the greater reactivity of the selenosemicarbazone, and glutathione's key role in selenosemicarbazone and thiosemicarbazone anticancer activity.
Collapse
Affiliation(s)
- Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
| | - Stanley Zhang
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
| | - Xiao Zhao
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
| | - Tharushi P Wijesinghe
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
| | - Mediha Suleymanoglu
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34093, Turkey
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
| | - Paul V Bernhardt
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane 4072, Australia
| | - Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
10
|
Garemilla SSS, Gampa SC, Garimella S. Role of the tumor microenvironment in cancer therapy: unveiling new targets to overcome drug resistance. Med Oncol 2025; 42:202. [PMID: 40332723 DOI: 10.1007/s12032-025-02754-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: 09/11/2024] [Accepted: 04/28/2025] [Indexed: 05/08/2025]
Abstract
Cancer is a leading cause of death globally, with resistance to therapy representing a major obstacle to effective treatment. The tumor microenvironment (TME), comprising a complex network to cellular and non-cellular components including cancer-associated fibroblasts, immune cells, the extracellular matrix and region of hypoxia, is integral to cancer progression and therapeutic resistance. This review delves into the multifaceted interactions within the TME that contribute to tumor growth, survival and immune evasion. Key elements such as the role of cancer- associated fibroblasts in remodeling the extracellular matrix and promoting angiogenesis, the influence of immune cells such as tumor-associated macrophages in creating an immunosuppressive milieu and the impact of hypoxia conditions on metabolic adaptation and therapy resistance are thoroughly examined. This review evaluates current and emerging TME-targeted therapeutic strategies, including inhibitors of extracellular matrix components, modulators of immune cell activity and approached to alleviate hypoxia. Combination therapies that integrate TME-targeted agents with conventional treatments such as chemotherapy and immunotherapy are also discussed for their potential to enhance treatment efficacy and circumvent resistance mechanisms. By synthesising recent advances in TME research and therapeutic innovation, this paper aims to underscore the importance of TME in cancer therapy and highlight promising avenues for improving patient outcomes through targeted intervention.
Collapse
Affiliation(s)
| | - Siri Chandana Gampa
- Department of Life Sciences, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh, 530045, India
| | - Sireesha Garimella
- Department of Life Sciences, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh, 530045, India.
| |
Collapse
|
11
|
Wang Q, Zhang X, Han C, Lv Z, Zheng Y, Liu X, Du Z, Liu T, Xue D, Li T, Wang L. Immunodynamic axis of fibroblast-driven neutrophil infiltration in acute pancreatitis: NF-κB-HIF-1α-CXCL1. Cell Mol Biol Lett 2025; 30:57. [PMID: 40335899 DOI: 10.1186/s11658-025-00734-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Acute pancreatitis (AP) is a sterile inflammation, and 10-20% of cases can progress to severe acute pancreatitis (SAP), which seriously threatens human life and health. Neutrophils and their extracellular traps (NETs) play an important role in the progression of AP. However, the immunodynamic factors between the excessive infiltration of neutrophils during the occurrence of AP have not been fully elucidated. METHODS Adult male C57BL/6 J mice were selected. An AP model was induced by cerulein, and a control group was set up. Single-cell sequencing technology was used to reveal the cell atlas of AP pancreatitis tissue. In vivo, the model mice were treated with anti-Ly6G antibody, DNase I, SC75741, PX-478, and SRT3109 respectively. In vitro, human pancreatic stellate cells were treated with hypoxia, H2O2, NAC, and JSH-2, and co-cultured with neutrophils in Transwell chambers. The severity of inflammation was evaluated, and the molecular mechanism by which fibroblasts exacerbate AP was revealed through techniques such as cell colony formation assay, cell migration assay, cell transfection, immunofluorescence, flow cytometry, Western blot, reverse-transcription quantitative polymerase chain reaction (RT-qPCR), and co-immunoprecipitation (co-IP). RESULTS The study showed that the elimination of neutrophils and NETs could significantly improve AP. Single-cell RNA sequencing (scRNA-seq) indicated that both neutrophils and fibroblasts in pancreatic tissue exhibited heterogeneity during AP. Among them, neutrophils highly expressed CXCR2, and fibroblasts highly expressed CXCL1. Further experimental results demonstrated that the infiltration of neutrophils in the early stage of AP was related to the activation of fibroblasts. The activation of fibroblasts depended on the nuclear factor kappa B (NF-κB) signaling pathway induced by hypoxia. NF-κB enhanced the activation of pancreatic stellate cells (PSCs) and the secretion of CXCL1 by directly promoting the transcription of HIF-1α and indirectly inhibiting PHD2, resulting in the accumulation of HIF-1α protein. The NF-κB-HIF-1α signal promoted the secretion of CXCL1 by fibroblasts through glycolysis and induced the infiltration of neutrophils. Finally, blocking the NF-κB-HIF-1α-CXCL1 signaling axis in vivo reduced the infiltration of neutrophils and improved AP. CONCLUSIONS This study, for the first time, demonstrated that activation of fibroblasts is one of the immunological driving factors for neutrophil infiltration and elucidated that glycolysis driven by the NF-κB-HIF-1α pathway is the intrinsic molecular mechanism by which fibroblasts secrete CXCL1 to chemotactically attract neutrophils. This finding provides a highly promising target for the treatment of AP.
Collapse
Affiliation(s)
- Qiang Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiao Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Chenglong Han
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Zhenyi Lv
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yi Zheng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuxu Liu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiwei Du
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianming Liu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongbo Xue
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Tao Li
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China.
| | - Liyi Wang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
12
|
Esposito T, Pentimalli F, Giordano A, Cortellino S. Vitamins and dietary supplements in cancer treatment: is there a need for increased usage? Expert Rev Anticancer Ther 2025:1-24. [PMID: 40322898 DOI: 10.1080/14737140.2025.2501077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/29/2025] [Indexed: 05/08/2025]
Abstract
INTRODUCTION Vitamins are essential for homeostasis and proper functioning of organisms. These micronutrients prevent tumor onset by functioning as antioxidants and enzymatic cofactors involved in anti-stress and immune responses, modulating epigenetic regulators, and shaping the microbiota composition. Unbalanced diets and sedentary lifestyles contribute to obesity, associated with increasing cancer risk. Cancer patients often exhibit vitamin deficiencies due to chronic inflammation, anticancer therapies, and tumor-induced metabolic changes, leading to malnutrition and cachexia. AREAS COVERED This review critically analyzes preclinical and clinical studies, sourced from PubMed and ClinicalTrials.gov databases, that investigate the potential benefits of vitamin supplementation and dietary interventions, such as intermittent fasting and ketogenic diets, in mouse tumor models and cancer patients. This analysis elucidates the limitations of such interventions and suggests optimal dietary strategies to prevent cancer and enhance patients' quality of life and prognosis. EXPERT OPINION To date, clinical studies have found no substantial benefit of over-the-counter vitamin supplements and dietary interventions on cancer patients' health and prognosis. To prevent the spread of useless and potentially harmful products by the nutraceutical industry, establishing a regulatory authority is necessary to monitor and ensure product quality and validity before commercialization.
Collapse
Affiliation(s)
- Teresa Esposito
- Department of Clinical Dietetics and Metabolic Diseases, Cavalier Raffaele Apicella Hospital, ASL Napoli 3 Sud, Naples, Italy
| | - Francesca Pentimalli
- Department of Medicine and Surgery, LUM University "Giuseppe De Gennaro", Bari, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Salvatore Cortellino
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, Naples, Italy
- S.H.R.O. Italia Foundation ETS, Turin, Italy
| |
Collapse
|
13
|
Dreyer SB, Beer P, Hingorani SR, Biankin AV. Improving outcomes of patients with pancreatic cancer. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01019-9. [PMID: 40329051 DOI: 10.1038/s41571-025-01019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
14
|
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. [PMID: 40326636 DOI: 10.1021/acsnano.4c16628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
15
|
Neophytou C, Stylianopoulos T, Mpekris F. The synergistic potential of mechanotherapy and sonopermeation to enhance cancer treatment effectiveness. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:13. [PMID: 40337117 PMCID: PMC12052595 DOI: 10.1038/s44341-025-00017-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/04/2025] [Indexed: 05/09/2025]
Abstract
Inefficient drug delivery in tumors, especially in desmoplastic cancers, arises from blood vessel collapse due to tumor stiffening and mechanical compression. Vessel collapse also leads to hypoxia, immune evasion, and metastasis, reducing treatment efficacy. Mechanotherapeutics and ultrasound sonopermeation, which address tumor stiffness and enhance vessel permeability, respectively, show promise in restoring tumor microenvironment abnormalities and improving drug delivery. This perspective highlights their independent and combined potential to optimize cancer therapy.
Collapse
Affiliation(s)
- Constantina Neophytou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
16
|
Gao Y, Chen Q, Wu Z, Yuan L. Regulation of pancreatic β cells by exosomes from different sources. Diabetes Res Clin Pract 2025:112222. [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] [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
|
17
|
Lei W, Zhou K, Lei Y, Li Q, Zhu H. Pathogenesis and Systemic Treatment of Hepatocellular Carcinoma: Current Status and Prospects. Mol Cancer Ther 2025; 24:692-708. [PMID: 39417575 DOI: 10.1158/1535-7163.mct-24-0403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/14/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
Hepatocellular carcinoma (HCC) remains one of the major threats to human health worldwide. The emergence of systemic therapeutic options has greatly improved the prognosis of patients with HCC, particularly those with advanced stages of the disease. In this review, we discussed the pathogenesis of HCC, genetic alterations associated with the development of HCC, and alterations in the tumor immune microenvironment. Then, important indicators and emerging technologies related to the diagnosis of HCC are summarized. Also, we reviewed the major advances in treatments for HCC, offering insights into future prospects for next-generation managements.
Collapse
Affiliation(s)
- Wanting Lei
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kexun Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ye Lei
- College of Liberal Arts, Neijiang Normal University, Neijiang, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Oñate MK, Oon C, Bhattacharyya S, Low V, Chen C, Zhao X, Arnold F, Yan Z, Pramod S, Hang Y, Ho YJ, Lowe SW, Kim SK, Xia Z, Sherman MH. Stromal KITL/SCF Maintains Pancreas Tissue Homeostasis and Restrains Tumor Progression. Cancer Discov 2025; 15:913-929. [PMID: 39918337 PMCID: PMC12046321 DOI: 10.1158/2159-8290.cd-24-1079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/15/2024] [Accepted: 02/05/2025] [Indexed: 02/12/2025]
Abstract
SIGNIFICANCE By analyzing transcriptional programs in healthy and tumor-associated pancreatic mesenchyme, we find that a subpopulation of mesenchymal cells in healthy pancreas tissue expresses the paracrine signaling factor KITL. The loss of mesenchymal KITL is an accompanying and permissive feature of pancreas tumor evolution, with potential implications for cancer interception. See related article by Dolskii and Cukierman, p. 872.
Collapse
Affiliation(s)
- Maria Kathrina Oñate
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Chet Oon
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Sohinee Bhattacharyya
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Vivien Low
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Canping Chen
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Xiaofan Zhao
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Frank Arnold
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Ziqiao Yan
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
| | - Sneha Pramod
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Graduate School of Medical Science, Weill Cornell Medicine, New York, New York
| | - Yan Hang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, California
| | - Yu-Jui Ho
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Scott W. Lowe
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Seung K. Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, California
- Department of Medicine, Stanford University School of Medicine, Stanford, California
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Zheng Xia
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Mara H. Sherman
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
19
|
Chang DH, Wang F, Palecek SP, Lynn DM. Slippery Liquid-Infused Porous Surfaces Infused with Thermotropic Liquid Crystals Enable Droplet-Based, Naked-Eye Reporting of Changes in Peptide Structure and Protease Activity. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40314309 DOI: 10.1021/acsami.5c02541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
We report the design of liquid crystal-infused "slippery" liquid-infused porous surfaces (LC-SLIPS) that permit naked-eye detection and reporting on the structural differences and activities of peptides and protease enzymes in aqueous media. We demonstrate that small (e.g., 20 μL) droplets of aqueous solutions placed in contact with LC-SLIPS exhibit sliding behaviors that vary substantially with the concentrations, structures, and physicochemical properties (e.g., hydrophobicity) of model amphiphilic β- and α/β-peptides dissolved within them. These large differences in sliding times permit naked-eye detection and discrimination of changes in peptide structure, including side-chain substitution, end group structure, backbone structure, and charge that correlate with differences in peptide amphiphilicity. We demonstrate further that LC-SLIPS can be used to monitor other biochemical processes, including digestion by proteases, that affect changes in the structures of amphiphilic peptides and can, thus, be used to develop novel, naked-eye assays that can report sensitively on enzymatic activity. As proof of concept, we show that large and visually observable changes in droplet sliding resulting from the degradation of a model peptide can be used to detect the presence of trypsin in aqueous solutions at levels as low as 12.5 ng/mL. That result, in turn, served as the basis of an LC-SLIPS-based assay that can be used to detect clinically relevant concentrations (from 25 to 25,000 ng/mL) of trypsinogen, a well-established biomarker for acute pancreatitis, in samples of synthetic urine. This "sliding" assay is conceptually straightforward and requires only visual monitoring and/or a hand-held stopwatch for readout, highlighting the potential for low-cost, point-of-care diagnostics applications. Overall, our results demonstrate the ability of LC-SLIPS to capture and report structural information relevant to other therapeutic properties and applications of amphiphilic peptides that could also be useful in the context of drug design and screening.
Collapse
Affiliation(s)
- Douglas H Chang
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Dr., Madison, Wisconsin 53706, United States
| | - Fengrui Wang
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Ave., Madison, Wisconsin 53706, United States
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Dr., Madison, Wisconsin 53706, United States
| | - David M Lynn
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Dr., Madison, Wisconsin 53706, United States
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Ave., Madison, Wisconsin 53706, United States
| |
Collapse
|
20
|
Kim DU, Kweon B, Oh JY, Noh GR, Lim Y, Yu J, Kim MJ, Kim DG, Park SJ, Bae GS. Curcumin ameliorates cerulein‑induced chronic pancreatitis through Nrf‑2/HO‑1 signaling. Mol Med Rep 2025; 31:136. [PMID: 40145554 PMCID: PMC11963747 DOI: 10.3892/mmr.2025.13501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Chronic pancreatitis (CP) is an invasive inflammatory disorder characterized by endocrine and exocrine dysfunction. There are currently no effective drugs for the treatment of CP. The present study investigated whether curcumin improves cerulein‑induced CP fibrosis in a mouse model and pancreatic stellate cells (PSCs). The CP mouse model was established by intraperitoneally injecting cerulein (50 µg/kg) for 3 weeks (six times at 1 h intervals/day; 4 days/week). To investigate the effects of curcumin, dimethyl sulfoxide or curcumin was injected intraperitoneally 1 h before the first daily injection of cerulein. To determine the severity of CP, the pancreas was harvested 24 h after the last cerulein injection for histological examination and assessment of PSC activation and collagen deposition. Additionally, levels of the nuclear factor erythroid 2‑related factor 2 (Nrf2) and heme oxygenase‑1 (HO‑1) were evaluated to determine the mechanism underlying the anti‑fibrotic effect of curcumin in PSCs. Curcumin improved pancreatic injury associated with CP by inhibiting PSC activation and collagen deposition. Moreover, curcumin increased HO‑1 expression levels via the activation of Nrf2 in PSCs, which suppressed the activation of PSCs. In conclusion, the present results suggest that curcumin can ameliorate pancreatic fibrosis induced by repetitive cerulein challenges via the induction of HO‑1 and is a beneficial agent for the treatment of CP.
Collapse
Affiliation(s)
- Dong-Uk Kim
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Bitna Kweon
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Jin-Young Oh
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Gyeong-Ran Noh
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Yebin Lim
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Jihyun Yu
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Myoung-Jin Kim
- Department of Herbology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Dong-Gu Kim
- Department of Herbology, College of Korean Medicine, Dong-Eui University, Busan, Gyeongnam 47887, Republic of Korea
| | - Sung-Joo Park
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
- Department of Herbology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Gi-Sang Bae
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
- Research Center of Traditional Korean Medicine, Wonkwang University, Iksan-daero, Iksan 54538, Republic of Korea
| |
Collapse
|
21
|
Duhn J, von Fritsch L, Bolm L, Braun R, Honselmann K, Litkevych S, Kist M, Deichmann S, Tol KKV, Franke B, Reinwald F, Sackmann A, Holleczek B, Krauß A, Klinkhammer-Schalke M, Zeissig SR, Keck T, Wellner UF, Abdalla TSA. Perioperative and oncologic outcomes after total pancreatectomy and pancreatoduodenectomy for pancreatic head adenocarcinoma-A propensity score-matched analysis from the German Cancer Registry Group. Surgery 2025; 181:109292. [PMID: 40101369 DOI: 10.1016/j.surg.2025.109292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/08/2025] [Accepted: 02/01/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND To compare perioperative morbidity and mortality in patients receiving pancreatoduodenectomy or total pancreatectomy for pancreatic head adenocarcinoma using German Cancer Registry data. METHODS Anonymized pooled data were retrieved from regional cancer registries participating in the German Cancer Registry Group of the Association of German Tumor Centers. Included were patients diagnosed with pancreatic head adenocarcinoma since 2016, receiving curative intent pancreatoduodenectomy or total pancreatectomy. Patients were propensity-score matched according to age, sex, and histopathology. Primary endpoints were 30- and 90-day postoperative mortality. Secondary endpoints were administration of adjuvant chemotherapy, long-term survival, and patterns of cancer recurrence. The data were analyzed using R. RESULTS In total, 756 patients per treatment group were matched for further analyses. R0-resection rate was comparable between pancreatoduodenectomy and total pancreatectomy (69.6 vs 73.4%, P = .154). The 30-day (9.5 vs 4.8%, P < .001) and 90-day postoperative mortality (18.0 vs 11.0%, P < .001) rates were significantly lower after pancreatoduodenectomy compared with total pancreatectomy. After pancreatoduodenectomy, more patients received adjuvant chemotherapy (43.6 vs 53.3%, P < .001) and time to adjuvant chemotherapy was shorter (60.1 vs 52.7 days, P = .002) compared with total pancreatectomy. Long-term overall survival was worse after total pancreatectomy (P < .001), also in patients receiving adjuvant chemotherapy (P = .019). The sites of recurrence were comparable between both groups (P = .274). CONCLUSION The results of this study show greater perioperative morbidity and mortality after total pancreatectomy compared with pancreatoduodenectomy for pancreatic head malignancy. Also, long-term survival was worse after total pancreatectomy. These results emphasize the role of pancreatoduodenectomy as a standard surgical procedure for pancreatic head adenocarcinoma and suggest that total pancreatectomy should only be performed in selected patients.
Collapse
Affiliation(s)
- Jannis Duhn
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Lennart von Fritsch
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Louisa Bolm
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Rüdiger Braun
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Kim Honselmann
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Stanislav Litkevych
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Markus Kist
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Steffen Deichmann
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Kees Kleihues-van Tol
- Network for Care, Quality and Research in Oncology, German Cancer Registry Group of the Association of German Tumor Centers (ADT), Berlin, Germany
| | - Bianca Franke
- Network for Care, Quality and Research in Oncology, German Cancer Registry Group of the Association of German Tumor Centers (ADT), Berlin, Germany
| | - Fabian Reinwald
- Cancer Registry of Rhineland-Palatinate in the Institute for Digital Health Data, Mainz, Germany
| | - Andrea Sackmann
- Hessian Cancer Registry, Hessian Office for Health and Care, Frankfurt, Germany
| | | | - Anna Krauß
- Cancer Registry Mecklenburg-Western Pomerania, Greifswald, Germany
| | - Monika Klinkhammer-Schalke
- Network for Care, Quality and Research in Oncology, German Cancer Registry Group of the Association of German Tumor Centers (ADT), Berlin, Germany
| | - Sylke R Zeissig
- Network for Care, Quality and Research in Oncology, German Cancer Registry Group of the Association of German Tumor Centers (ADT), Berlin, Germany; Institute of Clinical Epidemiology and Biometry (ICE-B), University of Würzburg, Würzburg, Germany
| | - Tobias Keck
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.
| | - Ulrich F Wellner
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Thaer S A Abdalla
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| |
Collapse
|
22
|
Liang Z, Ye Y, Deng Z, Lan H, Liu C, Xu Y, Fan M, Liu Z, Wu P, An L, Wang C. CHPF2 as a novel biomarker and ponicidin as a potential therapeutic agent in hepatocellular carcinoma. Pharmacol Res 2025; 215:107698. [PMID: 40107635 DOI: 10.1016/j.phrs.2025.107698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
Hepatocellular carcinoma (HCC) was associated with high morbidity and mortality, representing a significant health challenge. Chondroitin sulfate (CS), a glycosaminoglycan composed of glucuronic acid and N-acetylgalactosamine, is implicated in HCC progression through its role in cancer cell migration and proliferation as well as interactions with cell surface receptors integrin β-1 and CD44. Chondroitin polymerization factor 2 (CHPF2), the key to CS synthesis, has an undefined role in HCC. Our study aims to demonstrate that decreasing CHPF2 enzyme activity can inhibit the migration and proliferation of HCC cells. Bioinformatics analysis and in vitro experiments on clinical HCC samples confirmed the knockdown of CHPF2 inhibited HCC cell proliferation and migration. We further explored Rabdosia rubescens, a plant used in cancer therapy, for its potential to modulate CHPF2. Structural biology and ligand fishing identified ponicidin, a compound that significantly suppresses HCC cell growth and migration in both in vitro and in vivo models. These findings propose CHPF2 as a novel biomarker and ponicidin as a potential therapeutic agent for HCC management.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/genetics
- Humans
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/genetics
- Cell Proliferation/drug effects
- Cell Movement/drug effects
- Animals
- Cell Line, Tumor
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
- Mice, Nude
- Male
- MARVEL Domain-Containing Proteins/metabolism
- MARVEL Domain-Containing Proteins/genetics
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Mice, Inbred BALB C
- Hep G2 Cells
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/therapeutic use
Collapse
Affiliation(s)
- Zuhui Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Yingyi Ye
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Zhihong Deng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Huan Lan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Caihong Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Yuanhang Xu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Minqi Fan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Zhongqiu Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Peng Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China.
| | - Lin An
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China.
| | - Caiyan Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China.
| |
Collapse
|
23
|
Shakiba M, Tuveson DA. Macrophages and fibroblasts as regulators of the immune response in pancreatic cancer. Nat Immunol 2025; 26:678-691. [PMID: 40263612 DOI: 10.1038/s41590-025-02134-6] [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: 10/14/2024] [Accepted: 03/13/2025] [Indexed: 04/24/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the few cancers that has yet to benefit from immunotherapies. This is primarily a result of its characteristic 'cold' tumor microenvironment composed of cancer-associated fibroblasts (CAFs), a dense network of extracellular matrix and several immune cell types, the most abundant of which are the tumor-associated macrophages (TAMs). Advances in single-cell and spatial technologies have elucidated the vast functional heterogeneity of CAFs and TAMs, their symbiotic relationship and their cooperative role in the tumor microenvironment. In this Review, we provide an overview of the heterogeneity of CAFs and TAMs, how they establish an immunosuppressive microenvironment and their collaboration in the remodeling of the extracellular matrix. Finally, we examine why the impact of immunotherapy in PDAC has been limited and how a detailed molecular and spatial understanding of the combined role of CAFs and TAMs is paramount to the design of effective therapies.
Collapse
Affiliation(s)
- Mojdeh Shakiba
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY, USA
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY, USA.
| |
Collapse
|
24
|
Kølle IS, Hesthaven AS, Davidsen L, Hagn-Meincke R, Drewes AM, Pedersen IS, Ejstrud P, Henriksen SD, Olesen SS. Diagnostic yield of second-line aetiological workup in patients with presumed idiopathic acute pancreatitis: a retrospective cohort study. Scand J Gastroenterol 2025; 60:485-493. [PMID: 40160120 DOI: 10.1080/00365521.2025.2485135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/14/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND After an aetiological (first-line) workup, the cause of acute pancreatitis remains unidentified in a significant proportion of cases, a condition known as idiopathic acute pancreatitis (IAP). METHODS Retrospective cohort study involving patients with presumed IAP referred for second-line aetiological workup. The completion of first-line aetiological evaluations was assessed upon referral, and the diagnostic outcomes of second-line investigations were evaluated. Over a one-year follow-up period, we documented acute pancreatitis recurrence and patient mortality. Recurrence risk was analysed using an age-adjusted Cox regression model, stratified by treatable versus non-treatable aetiologies. RESULTS We identified 161 patients with presumed IAP, among whom 81 (50%) had recurrent acute pancreatitis. In total, 115 patients (71%) had a complete first-line aetiological workup. The overall diagnostic yield of the second-line aetiological workup was 25% (95% confidence interval [CI] 18-32%). Among second-line tests, the highest diagnostic yield was found for endoscopic ultrasound (34%, 95% CI 20-50%) and genetic testing (37%, 95% CI 22-53%). The most frequent aetiologies identified were biliary pancreatitis (16 patients [10%]) and pancreatitis with a genetic mutation (15 patients [9%]). Neoplasia was identified in two patients. A treatable aetiology was associated with a numerically reduced pancreatitis recurrence risk (Hazard Ratio 0.50, 95% CI 0.07-3.85, p = 0.51). No patient died during the follow-up period. CONCLUSION A second-line aetiological workup can identify the aetiology in 25% of patients with presumed IAP. The most frequent aetiologies are biliary pancreatitis and pancreatitis with a genetic mutation.
Collapse
Affiliation(s)
- Ida Saksenborg Kølle
- Centre for Pancreatic Diseases and Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Andreas Svenstrup Hesthaven
- Centre for Pancreatic Diseases and Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Line Davidsen
- Centre for Pancreatic Diseases and Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Rasmus Hagn-Meincke
- Centre for Pancreatic Diseases and Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Asbjørn Mohr Drewes
- Centre for Pancreatic Diseases and Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Inge Søkilde Pedersen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Molecular Diagnostics, Aalborg University Hospital, Aalborg, Denmark
| | - Per Ejstrud
- Department of Gastrointestinal Surgery, Aalborg University Hospital, Aalborg, Denmark
| | - Stine Dam Henriksen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Gastrointestinal Surgery, Aalborg University Hospital, Aalborg, Denmark
| | - Søren Schou Olesen
- Centre for Pancreatic Diseases and Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
25
|
Abu-Serie MM, Gutiérrez-García AK, Enman M, Vaish U, Fatima H, Dudeja V. Ferroptosis- and stemness inhibition-mediated therapeutic potency of ferrous oxide nanoparticles-diethyldithiocarbamate using a co-spheroid 3D model of pancreatic cancer. J Gastroenterol 2025; 60:641-657. [PMID: 39888413 PMCID: PMC12014774 DOI: 10.1007/s00535-025-02213-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/10/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with a high mortality rate and exhibits a limited response to apoptosis-dependent chemotherapeutic drugs (e.g., gemcitabine, Gem). This is mainly attributed to the antioxidant defense system (glutathione and aldehyde dehydrogenase (ALDH) 1A1), which sustains stemness features of cancer stem cells (CSCs) and activated pancreatic stellate cells (PSCs)-generated excess stromal proteins. This dense stroma retards drug delivery. METHODS This study established co-spheroid model consisting of mouse PDAC cell line (KPC) and PSCs (1:5) to accurately investigate the anti-PDAC activity of nanocomplex of ferrous oxide nanoparticles-diethyldithiocarbamate (FeO NPs-DE), compared to Gem, using in vitro and in vivo 3D models. RESULTS In vitro and in vivo co-spheroid models demonstrated higher therapeutic efficacy of FeO NPs-DE than Gem. FeO NPs-DE induced selective accumulation of iron-dependent ferroptosis (non-apoptosis)-generated a lethal lipid peroxidation that was potentiated by DE-mediated glutathione and ALDH1A1 suppression. This led to collapse of stemness, as evidenced by down-regulating CSC genes and p-AKT protein expression. Subsequently, gene and/or protein levels of PSC activators (transforming growth factor (TGF)-β, plasminogen activator inhibitor-1, ZEB1, and phosphorylated extracellular signal-regulated kinase) and stromal proteins (collagen 1A2, smooth muscle actin, fibronectin, and matrix metalloproteinase-9) were suppressed. Moreover, DE of nanocomplex enhanced caspase 3-dependent apoptosis with diminishing the main oncogene, BCL-2. CONCLUSIONS FeO NPs-DE had a stronger eradicating effect than Gem on primary and metastatic peritoneal PDAC tumors. This nanocomplex-mediated ferroptosis and stemness inhibition provides an effective therapeutic approach for PDAC.
Collapse
Affiliation(s)
- Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El‑Arab City, Alexandria, 21934, Egypt.
| | - Ana K Gutiérrez-García
- Division of Surgical Oncology, Department of Surgery, University of Alabama at Birmingham (UAB), Birmingham, Alabama, 35294, USA
| | - Macie Enman
- Division of Surgical Oncology, Department of Surgery, University of Alabama at Birmingham (UAB), Birmingham, Alabama, 35294, USA
| | - Utpreksha Vaish
- Division of Surgical Oncology, Department of Surgery, University of Alabama at Birmingham (UAB), Birmingham, Alabama, 35294, USA
| | - Huma Fatima
- Department of Pathology, Division of Anatomic Pathology, University of Alabama at Birmingham (UAB), Birmingham, Alabama, 35249, USA
| | - Vikas Dudeja
- Division of Surgical Oncology, Department of Surgery, University of Alabama at Birmingham (UAB), Birmingham, Alabama, 35294, USA
| |
Collapse
|
26
|
Duggirala S, Balasubramanian V, Seetharaman A, Murugan S, Roy J, Hassan S, V DR, Venkatraman G, Rayala SK. Transcriptome Analysis of Human Pancreatic Stellate Cells Co-cultured With PAK1-Modulated Cells Revealed the Role of Cytokine Pathway in Tumor Microenvironment. Pancreas 2025; 54:e414-e422. [PMID: 40314739 DOI: 10.1097/mpa.0000000000002450] [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: 06/28/2024] [Accepted: 11/26/2024] [Indexed: 05/03/2025]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with high metastatic ability, poor prognosis, and resistant to treatment. Tumor microenvironment plays a major role in the complexity of PDAC. OBJECTIVE OF THE STUDY The aim of the study was to examine the role of P21 activated kinase-1 (PAK1) in the sustenance of tumor microenvironment to enable tumor growth and progression. METHODOLOGY The effect of PAK1 in the tumor microenvironment was analyzed using a novel co-culture method involving pancreatic cancer cells and pancreatic stellate cells. The 2 cell types were grown in both direct and indirect cell culture models to facilitate the juxtracrine signaling and establish a secretome network. The established network was studied using the transcriptome sequencing of PAK1-modulated MIA PaCa-2 cells co-cultured with stellate cells. RESULTS The results showed that PAK1 influenced cells have increased interferon pathway when compared to PAK1 depleted cells. The levels of chemokine CCL3 was altered in PAK1-modulated cells as evidenced by the bioinformatic, QPCR, and ELISA analysis. The pathway and interactome analysis showed that CCL3 promotes interferon activation and myofibroblast differentiation in pancreatic cancer microenvironment. These results might help in identifying the PAK1 induced metastatic network in pancreatic cancer. Further investigation will provide adequate evidence of CCL3 and PAK1 in pancreatic carcinogenesis and metastasis. CONCLUSIONS The present study provides an understanding of tumor microenvironment and involvement of inflammatory cytokines in a juxtacrine mechanism to aggravate and accelerate pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Sridevi Duggirala
- Department of Biotechnology, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu, India
- Department of Cancer Biology & Molecular Diagnostics, Cancer Institute (W.I.A), Chennai, India
| | - Vaishnavi Balasubramanian
- Department of Human Genetics, Sri Ramachandra Institute of Higher education and Research, Porur, Chennai, India
| | - Abirami Seetharaman
- Department of Biotechnology, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu, India
| | - Sowmiya Murugan
- Department of Biotechnology, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu, India
| | - Joydeep Roy
- Department of Biotechnology, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu, India
| | | | - Devi Rajeswari V
- Department of Bio-Medical Sciences, School of Bio Sciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Ganesh Venkatraman
- Department of Bio-Medical Sciences, School of Bio Sciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Suresh Kumar Rayala
- Department of Biotechnology, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu, India
| |
Collapse
|
27
|
Liu Y, Xue R. Pancreatic stellate cell: Update on molecular investigations and clinical translation in pancreatic cancer. Int J Cancer 2025; 156:1672-1685. [PMID: 39825771 DOI: 10.1002/ijc.35326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025]
Abstract
Pancreatic cancer is a particularly aggressive tumor, distinguished by the presence of a prominent collagenous stroma and desmoplasia that envelops the tumor cells. Pancreatic stellate cell (PSC) contributes to the formation of a dense fibrotic stroma and has been demonstrated to facilitate tumor progression. As the significance of PSCs is increasingly revealed, more explorations are focused on the complex molecular mechanisms and tumor-stromal crosstalk in order to guide potential therapeutic approaches through deactivating or reprogramming PSCs. Nevertheless, significant challenges persist in translating preclinical discoveries into clinical applications. In this review, we expect to offer a comprehensive overview of the latest molecular advancements in PSCs, along with new insights into the clinical therapeutic strategies targeting PSCs.
Collapse
Affiliation(s)
- Yawei Liu
- School of Basic Medicine Sciences, Capital Medical University, Beijing, China
- Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Ran Xue
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
28
|
Jelleschitz J, Heider S, Kehm R, Baumgarten P, Ott C, Schnell V, Grune T, Höhn A. Insulitis and aging: Immune cell dynamics in Langerhans islets. Redox Biol 2025; 82:103587. [PMID: 40101534 PMCID: PMC11957801 DOI: 10.1016/j.redox.2025.103587] [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: 12/05/2024] [Revised: 02/07/2025] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
With increasing age, the risk for age-related type-2-diabetes also increases due to impaired glucose tolerance and insulin secretion. This disease process may be influenced by various factors, including immune cell triggered inflammation and fibrosis. Although immune cells are a necessary component of islets, little is known about immune cell accumulation, immune cell subtype shifts and subsequent influence on glucose metabolism in healthy aging. However, this is critical for understanding the mechanisms that influence β-cell health. Therefore, we studied young and old male C57BL/6J mice, focusing on immune cell composition, patterns of accumulation, and the presence of fibrosis within the pancreatic islets. Our findings demonstrate that insulitis occurs in healthy aged mice without immediate development of a diabetic phenotype. Aged islets exhibited an increase in leukocytes and a shift in immune cell composition. While insulitis typically involves excessive immune cell accumulation, we observed a moderate increase in macrophages and T-cells during aging, which may support β-cell proliferation via cytokine secretion. In fact, aged mice in our study showed an increase in β-cell mass as well as a partially higher insulin secretory capacity, which compensated for the loss of β-cell functionality in insulitic islets and led to improved glucose tolerance. Furthermore, fibrosis which is normally triggered by immune cells, increased with age but appears to reach a steady state, emphasizing the importance of counter-regulatory mechanisms and immune system regulation. Our results suggest, that immune cell subtypes change with age and that non-pathological accumulation of immune-cells may regulate glucose metabolism through secretion of cytokines.
Collapse
Affiliation(s)
- Julia Jelleschitz
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Sophie Heider
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Richard Kehm
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Patricia Baumgarten
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner site Berlin, Berlin, Germany
| | - Christiane Ott
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner site Berlin, Berlin, Germany
| | - Vanessa Schnell
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany; DZHK (German Center for Cardiovascular Research), Partner site Berlin, Berlin, Germany
| | - Annika Höhn
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany.
| |
Collapse
|
29
|
Ramai D, Collins B, Ofosu A, Mohan BP, Jagannath S, Tabibian JH, Girotra M, Barakat MT. Deep Learning Methods in the Imaging of Hepatic and Pancreaticobiliary Diseases. J Clin Gastroenterol 2025; 59:405-411. [PMID: 40193287 DOI: 10.1097/mcg.0000000000002125] [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: 04/09/2025]
Abstract
Reports indicate a growing role for artificial intelligence (AI) in the evaluation of pancreaticobiliary and hepatic conditions. A key focus is differentiating between benign and malignant lesions, which is crucial for treatment decisions. AI improves diagnostic accuracy through high sensitivity and specificity, while CNN algorithms enhance image analysis and reduce variability. These advancements aim to match the accuracy of pathologists in cancer detection. In addition, AI aids in identifying diagnostic markers, as early detection is essential. This article reviews the applications of machine learning and deep learning in the diagnosis of hepatic and pancreaticobiliary diseases. Although the use of AI in these specialized areas of gastroenterology is primarily confined to experimental trials, current models demonstrate significant potential for enhancing the detection, evaluation, and treatment planning of hepatic and pancreaticobiliary conditions.
Collapse
Affiliation(s)
- Daryl Ramai
- Division of Gastroenterology and Hepatology, University of Utah, Salt Lake City, UT
| | - Brendan Collins
- Division of Digestive Diseases, University of Cincinnati, Cincinnati, OH
| | - Andrew Ofosu
- Division of Digestive Diseases, University of Cincinnati, Cincinnati, OH
| | - Babu P Mohan
- Division of Gastroenterology and Hepatology, University of Utah, Salt Lake City, UT
| | - Soumya Jagannath
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - James H Tabibian
- Division of Gastroenterology, Olive View-UCLA Medical Center, Sylmar
- David Geffen School of Medicine at UCLA, Los Angeles
| | - Mohit Girotra
- Digestive Health Institute, Swedish Medical Center, Seattle, WA
| | | |
Collapse
|
30
|
Hubert MO, Mayerle J, Sirtl S. [Pancreatitis from drugs-Drugs for treatment of pancreatitis]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2025; 66:524-532. [PMID: 40298970 DOI: 10.1007/s00108-025-01888-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/04/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Drugs are a rare but important cause of acute pancreatitis (AP) due to potential therapeutic consequences, accounting for up to 5% of all cases of AP. The diagnosis of drug-induced AP is challenging due to mostly weak evidence and complex diagnostic criteria. OBJECTIVE This review article defines drug-induced AP, summarizes the evidence for drugs associated with AP and highlights the challenges in the diagnosis of this condition. The second part of the article focuses on the main pillars of AP treatment. CURRENT DATA The association of most drugs associated with AP is based on case reports and case series but there are no high-quality studies. There is sufficient evidence of a causal relationship for only 40 of more than 500 drugs associated with AP and for almost none of the drugs the causal mechanism has been definitively clarified. Several classification systems and criteria have been proposed to assess whether a drug causally triggers AP, with criteria including the temporal association, the exclusion of other causes and recurrence of AP after re-exposure. CONCLUSION The diagnosis of drug-induced AP remains a challenge, with many common drugs being incorrectly associated with AP. There is an urgent need for the development of biomarkers to facilitate the diagnosis of drug-induced AP. Drug treatment for AP is still primarily a needs-based fluid management and efficient analgesia. New and causal therapeutic approaches need clinical validation.
Collapse
Affiliation(s)
- Max Ole Hubert
- Medizinische Klinik und Poliklinik II, LMU Klinikum München, Marchioninistr. 15, 81377, München, Deutschland
| | - Julia Mayerle
- Medizinische Klinik und Poliklinik II, LMU Klinikum München, Marchioninistr. 15, 81377, München, Deutschland.
| | - Simon Sirtl
- Medizinische Klinik und Poliklinik II, LMU Klinikum München, Marchioninistr. 15, 81377, München, Deutschland
| |
Collapse
|
31
|
Azeez A, Noel C. Global status of research on quality of life in pancreatic cancer patients: A bibliometric and network analysis from 2005-2024. Clin Res Hepatol Gastroenterol 2025; 49:102595. [PMID: 40210107 DOI: 10.1016/j.clinre.2025.102595] [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: 12/21/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Pancreatic cancer (PC) is a major global health challenge, with rising incidence and mortality rates, particularly in high-socio-demographic index regions. Given its high mortality and significant morbidity, research on patient quality of life (QoL) has gained momentum, addressing symptom burdens, psychological distress, and treatment-related outcomes. Bibliometric analysis provides a valuable approach to mapping research trends, identifying key contributors, and forecasting future directions. OBJECTIVE This study aimed to map global research on QoL in pancreatic cancer patients, highlighting key findings, challenges, and future directions through bibliometric analysis over the past two decades. METHODS Data for this study were collected from the Web of Science Core Collection (WoSCC) database, using specific search strategies to retrieve relevant documents on the quality of life in pancreatic cancer patients. The data were analysed using the Bibliometrix R package to create knowledge maps and visualize research trends, collaborations, and emerging hotspots in the field. RESULTS A total of 819 articles on pancreatic cancer and quality of life were identified, with an average citation count of 47.13 per article, highlighting moderate academic impact. The research revealed a growing trend in collaborative efforts, with an average of 9.42 co-authors per article and 16 % international collaborations. The United States emerged as the leading contributor, with 203 publications and the highest citation count, followed by France and the United Kingdom. CONCLUSION This bibliometric analysis highlights the growing volume of pancreatic cancer and quality of life research, with a steady annual growth rate of 6.9 % and increasing collaboration, especially from the United States. However, despite the rising number of publications, a decline in citation impact for recent studies suggests a need for continued innovation in therapeutic strategies to improve clinical outcomes.
Collapse
Affiliation(s)
- Adeboye Azeez
- Gastrointestinal Research Unit, Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa.
| | - Colin Noel
- Gastrointestinal Research Unit, Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| |
Collapse
|
32
|
Yao GW, Li CX, Fan YX, Zhuo YZ, Zhang SK, Cui LH. Chaihu Guizhi Ganjiang Decoction ameliorates chronic pancreatitis by modulating the SK1/S1P signaling pathway. J Nat Med 2025; 79:706-720. [PMID: 40178769 DOI: 10.1007/s11418-025-01901-x] [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: 12/06/2024] [Accepted: 03/14/2025] [Indexed: 04/05/2025]
Abstract
Chronic pancreatitis (CP) is a progressive disease characterized by injury on pancreatic acinar cells (PACs), ongoing fibrosis, and gradual loss of exocrine and endocrine functions. Sphingosine kinase 1 (SK1) expression is elevated in injured PACs, and its metabolite sphingosine-1-phosphate (S1P) promotes the activation of pancreatic stellate cell (PSC) through autophagy and pyroptosis. Chaihu Guizhi Ganjiang Decoction (CGGD), a traditional Chinese medicine is widely used in the clinical treatment of digestive diseases. However, whether CCGD affects the SK1/S1P axis and relieves pancreatic damage through this pathway remains unknown. In this study, CP rats were treated with CGGD, individually or in combination with S1P and SKI-178 for four weeks to assess the effect of CGGD on pancreatic injury, fibrosis, autophagy and pyroptosis. The results showed that SK1, S1P and S1PR2 levels were increased in the pancreatic tissues of CP rats, while CGGD reduced these levels. Treatment with S1P exacerbated histological damage, promoted fibrosis, accelerated autophagy, and induced pyroptosis. Conversely, SKI-178 suppressed these effects. Notably, CGGD mitigated histological damage, decreased serum amylase and lipase levels, and alleviated pancreatic fibrosis induced by S1P. Furthermore, CGGD downregulated autophagy and pyroptosis induced by S1P, exhibiting an effect comparable to SKI-178 in CP. In conclusion, CGGD ameliorates pancreatic damage by reducing fibrosis, inhibiting autophagy, and suppressing pyroptosis through the SK1/S1P axis.
Collapse
Affiliation(s)
- Guo-Wang Yao
- Department of Gastrointestinal Surgery, Hospital of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Cai-Xia Li
- Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin, 300100, China
| | - Yu-Xing Fan
- Graduate School, Tianjin Medical University, Tianjin, 300070, China
| | - Yu-Zhen Zhuo
- Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
| | - Shu-Kun Zhang
- Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China
| | - Li-Hua Cui
- Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China.
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin, 300100, China.
- Institute of Acute Abdominal Diseases of Integrated Traditional Chinese and Western Medicine, Tianjin Nankai Hospital, 6 Changjiang Road, Tianjin, 300100, China.
| |
Collapse
|
33
|
Arseneau RJ, Kempster E, Bekkers C, Samson T, Gala-Lopez BL, Ramjeesingh R, Boudreau JE, Arnason T. Claudin 18 (43-14A clone) expression in pancreatic ductal adenocarcinoma: Assessment of a potential clinical biomarker for zolbetuximab therapy. Transl Oncol 2025; 55:102362. [PMID: 40117781 PMCID: PMC11979426 DOI: 10.1016/j.tranon.2025.102362] [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: 10/07/2024] [Revised: 03/08/2025] [Accepted: 03/16/2025] [Indexed: 03/23/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains highly lethal, with a five-year survival rate below 15 %. Claudin 18.2 (CLDN18.2) has emerged as a novel potential therapeutic target in PDAC. Zolbetuximab, a monoclonal antibody targeting CLDN18.2, has demonstrated therapeutic benefit in gastric cancers and is now in phase 2 clinical trials for PDAC. Trial eligibility for zolbetuximab requires tumor membranous immunohistochemical staining with the pan-claudin 18 companion diagnostic antibody clone 43-14A. However, the expression of CLDN18 detected using this clone has only been evaluated in 62 patients from a single retrospective study. Herein, we report immunohistochemical staining using 43-14A on surgically resected PDAC samples (n = 120). Samples were stained following the protocol used in clinical trials, using the 43-14A VENTANA antibody in a prediluted kit, and according to the manufacturer's recommended protocol. Positive cases were defined as ≥ 75 % of tumor cells exhibiting membranous staining with an intensity of ≥ 2+. Out of 120 PDAC cases, 39 (32.5 %) stained positive for CLDN18 with 43-14A. A significant association was observed between lower tumor grade and higher 43-14A staining (p < 0.05). CLDN18-positive cases demonstrated significantly improved survival at the cohort's median overall survival (23 months, p < 0.05), suggesting that claudin expression could serve as a both a diagnostic and prognostic marker. Our findings indicate that 32.5 % of PDAC tumors in this cohort are positive for CLDN18, suggesting that a significant proportion of patients with PDAC could benefit from zolbetuximab and other CLDN18.2 targeted immunotherapies if pancreatic cancer therapeutic trials prove successful.
Collapse
Affiliation(s)
- Riley J Arseneau
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | | | | | | | - Boris L Gala-Lopez
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada; Nova Scotia Health, Halifax, NS, Canada; Department of Microbiology & Immunology, Dalhousie University, NS, Canada
| | - Ravi Ramjeesingh
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada; Nova Scotia Health, Halifax, NS, Canada
| | - Jeanette E Boudreau
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada; Nova Scotia Health, Halifax, NS, Canada; Department of Microbiology & Immunology, Dalhousie University, NS, Canada
| | - Thomas Arnason
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Nova Scotia Health, Halifax, NS, Canada.
| |
Collapse
|
34
|
Lu S, Zhou Q, Zhao R, Xie L, Cao WM, Feng YX. Unraveling UPR-mediated intercellular crosstalk: Implications for immunotherapy resistance mechanisms. Cancer Lett 2025; 617:217613. [PMID: 40054654 DOI: 10.1016/j.canlet.2025.217613] [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: 11/20/2024] [Revised: 02/10/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Endoplasmic reticulum (ER) is the critical organelle that regulates essential cellular processes, including protein synthesis, folding, and post-translational modification, as well as lipid metabolism and calcium homeostasis. Disruption in ER homeostasis leads to a condition known as ER stress, characterized by the accumulation of misfolded or unfolded proteins. This triggers the unfolded protein response (UPR), an adaptive pathway mediated by three ER-resident sensors: inositol-requiring enzyme 1α (IRE1α), protein kinase R-like ER kinase (PERK), and activating transcription factor 6 (ATF6). Increasing evidence highlights sustained UPR activation in malignant and immune cells within the tumor microenvironment (TME), which promotes tumor progression and metastasis while simultaneously impairing antitumor immunity. This review explores how UPR-driven intercellular signaling influences immunotherapy resistance, focusing on the alterations occurring in tumor cells as well as in the surrounding immune environment. By providing insights into these mechanisms, we aim to highlight the therapeutic potential of targeting the UPR pathways in modulating cancer immunity.
Collapse
Affiliation(s)
- Si Lu
- Department of Head and Neck Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China; Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou, China
| | - Qimin Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rongjie Zhao
- Department of Gynecological Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, China
| | - Lei Xie
- Department of Head and Neck Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Wen-Ming Cao
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China.
| | - Yu-Xiong Feng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, China; Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou, China.
| |
Collapse
|
35
|
Li Z, Ren H, Zhang S, Sun C, Li Z, Niu P, Fei H, Xing C, Shi S, Zhao D. PD-L1 levels, TP53 mutation profiles, and survival outcomes in pancreatic cancer differ by immune-nutritional status. World J Surg Oncol 2025; 23:174. [PMID: 40301983 PMCID: PMC12042528 DOI: 10.1186/s12957-025-03818-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 04/15/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) frequently exhibits an immunosuppressive microenvironment coupled with malnutrition status. These features are instrumental in clinical management strategies for PDAC. METHODS Immune-nutrition status of patients was evaluated by integrating systemic immune-inflammatory index (SII) and prognostic nutritional index (PNI). Individuals were divided into SII-PNI Status positive (SPS+) group and SPS negative (SPS-) group. Morphology of tissues was evaluated by hematoxylin-eosin (H&E) staining. Expression of PD-L1 and p53 was detected using immunohistochemistry (IHC). RESULTS In this study, 530 eligible patients (mean ± SD age, 60.5 ± 9.17 years, 296 males [55.8%], 74 SPS+ [14.0%]) were included. These patients exhibited a median survival of 24 months (1-, 3- and 5-year survival rate; 72.9%, 34.7% and 25.1%, respectively). In the multivariate analysis, independent indicators for outcomes were identified as tumor size, lymph node metastasis and SPS (all p <.01). After matching and adjusting, patients with SPS+ exhibited a notably reduced overall survival compared to those with SPS- (14 vs. 25 months, p <.001), with hazard ratio (95% CI) of 1.79 (1.25-2.56). IHC revealed markedly elevated positive cell proportion of PD-L1 in SPS+ group (p <.01) and distinct p53 mutation patterns between SPS+ and SPS- groups (p =.03). Morphology demonstrated a dissimilar trend of differentiation levels between the two groups (p =.08). CONCLUSION The findings suggest poorer outcome, higher PD-L1 expression and distinct p53 mutation status of patients with SPS+. These patterns may contribute to PDAC management and strategic deployment of immunotherapy and targeted therapy.
Collapse
Affiliation(s)
- Zheng Li
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/ National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Hu Ren
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/ National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Shihui Zhang
- Department of Pathology, National Cancer Center, National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chongyuan Sun
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/ National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Zefeng Li
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/ National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Penghui Niu
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/ National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - He Fei
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/ National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Cheng Xing
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P.R. China.
| | - Susheng Shi
- Department of Pathology, National Cancer Center, National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Dongbing Zhao
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/ National Clinical Research for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
36
|
Fan H, Zhao H, Gao L, Dong Y, Zhang P, Yu P, Ji Y, Chen ZS, Liang X, Chen Y. CCN1 Enhances Tumor Immunosuppression through Collagen-Mediated Chemokine Secretion in Pancreatic Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500589. [PMID: 40287974 DOI: 10.1002/advs.202500589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/03/2025] [Indexed: 04/29/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense, immunosuppressive tumor microenvironment (TME) that limits therapeutic efficacy. This study investigates the role of cellular communication network factor 1 (CCN1, also known as Cyr61), an extracellular matrix-associated protein, in modulating the TME of PDAC. It is demonstrated that Ccn1 promotes PDAC progression by upregulating collagen and chemokine expression, thereby facilitating immune cell exclusion and enhancing tumor growth. Using a Ccn1-deficient PDAC model, decreased collagen and chemokine levels are observed, resulting in increased infiltration of cytotoxic immune cells and reduced myeloid-derived suppressor cells (MDSCs). Furthermore, Ccn1-deficient tumors exhibit heightened sensitivity to gemcitabine and show enhanced responsiveness to anti-programmed cell death 1 (anti-PD1) therapy. Mechanistically, Ccn1 regulates chemokine production through collagen expression, with chemokine levels remaining suppressed even upon interferon-gamma treatment in collagen-deficient cells. These findings highlight Ccn1 as a potential therapeutic target that reprograms the TME to enhance the efficacy of both chemotherapy and immunotherapy in PDAC, providing a novel approach for overcoming immune resistance in PDAC.
Collapse
Affiliation(s)
- Hongjie Fan
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Huzi Zhao
- Department of Pathology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, China
| | - Lili Gao
- Department of Pathology, Xinhua Hospital Affiliated to Medicine School of Shanghai Jiaotong University, Shanghai, 200082, China
| | - Yucheng Dong
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100006, China
| | - Pei Zhang
- Department of Mathematics, University of Maryland, College Park, Maryland, MD 20742, USA
| | - Pengfei Yu
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Yunfei Ji
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Xinmiao Liang
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Yang Chen
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| |
Collapse
|
37
|
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] [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
|
38
|
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] [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
|
39
|
Jin H, Zhang D, Ma Y, Meng L, Huang S, Su H, Xu J, Yao Y. YTHDC2 manipulates anti-tumoral macrophage polarization and predicts favorable outcomes in triple negative breast cancer. NPJ Precis Oncol 2025; 9:119. [PMID: 40274959 PMCID: PMC12022267 DOI: 10.1038/s41698-025-00880-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 03/11/2025] [Indexed: 04/26/2025] Open
Abstract
Triple-negative breast cancer (TNBC) possesses high malignant and metastatic rates among all subtypes. Chemotherapy is a standard of care for TNBC but only a small moiety of patients achieved complete relief (CR) after chemotherapy. The recent concept of tumor ecosystem has provided new insights into solutions from an approach of enhancing anti-tumoral immunity of macrophages. We hereby observed a positive correlation of YTHDC2 abundance with anti-tumoral gene markers of macrophages. YTHDC2-high macrophages also exerted interactions with other immune cells such as T helper cells, cytotoxic T cells, and NK cells. Further investigation on the transcriptional regulatory network identified six transcriptional factors upregulated by YTHDC2, and they together influenced the expressions of TWISTNB and the oncogene MYC. Additionally, our survival analysis prompted that YTHDC2 is prognostic of higher chemo-therapeutic efficacy and better survival outcomes. We demonstrated that ample macrophage YTHDC2 indicates anti-tumoral phenotype polarization and propitious survival outcome in post-treatment TNBC patients (Clinical trial registry name: Chinese Clinical Trial Registry, Registration No.: ChiCTR2400084513, Registration Date: 2024-05-20).
Collapse
Affiliation(s)
- Hao Jin
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Dongbo Zhang
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yufan Ma
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Lanlan Meng
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Cancer Pathogenesis and Precision Diagnosis and Treatment, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, China
| | - Songyin Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Hongjun Su
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jiannan Xu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yandan Yao
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province, China.
- Guangdong Provincial Key Laboratory of Cancer Pathogenesis and Precision Diagnosis and Treatment, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, China.
| |
Collapse
|
40
|
Yang Y, Qiang C, Jie Z, Ce H, Yan H, Xiu-Bin L, Wen-Mei F, Xu Z, Yu G. Exosomes derived from ccRCC cells confers fibroblasts activation to foster tumor progression through Warburg effect by downregulating PANK3. Cell Death Discov 2025; 11:198. [PMID: 40280913 PMCID: PMC12032068 DOI: 10.1038/s41420-025-02434-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/27/2024] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
The interaction between tumor-derived exosomes and stroma plays a crucial role in tumor progression. However, the mechanisms through which tumor cells influence stromal changes are not yet fully understood. In our study, through single-cell sequencing analysis of clear cell renal cell carcinoma tissues at varying stages of progression, we determined that the proportion of cancer-associated fibroblasts (CAFs) in advanced renal cell carcinoma tissues was notably higher compared to localized renal cell carcinoma tissues. Comparison of transcriptome sequencing and energy metabolism tests between CAFs primarily isolated from advanced renal cell carcinoma tissues and normal fibroblasts (NFs) revealed the occurrence of the Warburg effect during the fibroblast activation process. Additionally, we observed an increase in glucose transporter GLUT1 expression, total reactive oxygen species (ROS) levels, lactic acid production, and subsequent excretion of excess lactic acid through monocarboxylate transporter-4 (MCT4) in CAFs. Interestingly, renal cancer cells were found to uptake lactic acid via MCT1 upon interaction with CAFs, thereby enhancing their malignant phenotypes. Furthermore, the down-regulation of PANK3 induced by exosomes derived from renal cancer cells was identified as a crucial step in fibroblast activation. These findings indicate that exosomes play a role in facilitating intercellular communication between renal cancer cells and fibroblasts. Targeting this communication pathway could potentially offer new strategies for the prevention and treatment of advanced renal cell carcinoma.
Collapse
Affiliation(s)
- Yang Yang
- Department of Urology, the third Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Cheng Qiang
- Department of Urology, the third Medical Centre, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Zhu Jie
- Department of Urology, the third Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Han Ce
- Department of Urology, the third Medical Centre, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Huang Yan
- Department of Urology, the third Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Li Xiu-Bin
- Department of Urology, the third Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Fan Wen-Mei
- Department of Urology, the third Medical Centre, Chinese PLA General Hospital, Beijing, China.
| | - Zhang Xu
- Department of Urology, the third Medical Centre, Chinese PLA General Hospital, Beijing, China.
| | - Gao Yu
- Department of Urology, the third Medical Centre, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
41
|
Zhao B, Fang R, Schürmann H, Hemmer EJ, Mayer GL, Trajkovic-Arsic M, Althoff K, Yang J, Godfrey L, Liffers ST, Savvatakis K, Dorsch M, Grüner BM, Hahn S, Remke M, Lueong SS, Siveke JT. PLK1 blockade enhances the anti-tumor effect of MAPK inhibition in pancreatic ductal adenocarcinoma. Cell Rep 2025; 44:115541. [PMID: 40188436 DOI: 10.1016/j.celrep.2025.115541] [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: 01/19/2024] [Revised: 02/04/2025] [Accepted: 03/19/2025] [Indexed: 04/08/2025] Open
Abstract
Despite constitutive Ras/Raf/MAPK pathway activation in most pancreatic ductal adenocarcinomas (PDACs), treatment approaches targeting this pathway have primarily been unsuccessful. We conduct a drug library screen on an MEK inhibitor (MEKi)-resistant PDAC model and perform complementary pathway analysis to identify cellular resistance phenotypes. We use syngeneic models to investigate the molecular determinants of identified drug synergism. Our study reveals an enrichment for the hallmarks of G2/M checkpoints in MEKi-resistant phenotypes from all investigated models. We find overexpression of Polo-like kinase 1 (PLK1) and other G2/M checkpoint-related proteins in MEKi-resistant cells. We identify synergistic activity between MEK and PLK1 inhibition both in vitro and in vivo and mechanistically show that dual inhibition of the PLK1 and MEK pathways activates the JNK/c-JUN pathway. This causes the accumulation of DNA damage, ultimately leading to apoptotic cell death. Dual PLK1/MEK inhibition emerges as a promising targeted approach in PDAC.
Collapse
Affiliation(s)
- Ben Zhao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Rui Fang
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Hendrik Schürmann
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany; Department of Medical Oncology, West German Cancer Center, University Hospital Essen, 45147 Essen, Germany
| | - Erik Jan Hemmer
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Gina Lauren Mayer
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Marija Trajkovic-Arsic
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Kristina Althoff
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Jiajin Yang
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Laura Godfrey
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Sven T Liffers
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Konstantinos Savvatakis
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Madeleine Dorsch
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany; Department of Medical Oncology, West German Cancer Center, University Hospital Essen, 45147 Essen, Germany
| | - Barbara M Grüner
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany; Department of Medical Oncology, West German Cancer Center, University Hospital Essen, 45147 Essen, Germany
| | - Stephan Hahn
- Department of Molecular GI Oncology, Faculty of Medicine, Ruhr University Bochum, 44780 Bochum, Germany; Department of Internal Medicine, Ruhr University Bochum, Knappschaftskrankenhaus, 44780 Bochum, Germany
| | - Marc Remke
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, 40225 Düsseldorf, Germany; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada; Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 40225 Düsseldorf, Germany
| | - Smiths S Lueong
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany.
| | - Jens T Siveke
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany.
| |
Collapse
|
42
|
Liu Z, Ba Y, Shan D, Zhou X, Zuo A, Zhang Y, Xu H, Liu S, Liu B, Zhao Y, Weng S, Wang R, Deng J, Luo P, Cheng Q, Hu X, Yang S, Wang F, Han X. THBS2-producing matrix CAFs promote colorectal cancer progression and link to poor prognosis via the CD47-MAPK axis. Cell Rep 2025; 44:115555. [PMID: 40222008 DOI: 10.1016/j.celrep.2025.115555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/30/2024] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) display significant functional and molecular heterogeneity within the tumor microenvironment, playing diverse roles in cancer progression. Employing single-cell RNA sequencing data of colorectal cancer (CRC), we identified a subset of matrix CAFs (mCAFs) as a critical subtype that secretes THBS2, a molecule linked to advanced cancer stages and poor prognosis. Spatial transcriptomics and multiplex immunohistochemistry revealed clear spatial colocalization between THBS2-producing mCAFs and tumor cells. Mechanically, CAF-secreted THBS2 binds to CD47 on tumor cells, triggering the MAPK/ERK5 signaling pathway, which enhances tumor progression. The tumor-promoting role of THBS2 was further validated using fibroblast-specific THBS2 knockout mice, patient-derived organoids, and xenografts. Moreover, the transcription factor CREB3L1 was identified as a regulator of the transformation of normal fibroblasts into THBS2-producing mCAFs. These findings underscore the pivotal role of THBS2 in CRC progression and highlight the therapeutic potential of targeting the THBS2-CD47 axis and CREB3L1 in CRC.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China; Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Dan Shan
- Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YT, UK
| | - Xing Zhou
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Benyu Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yanan Zhao
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ruizhi Wang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jinhai Deng
- Richard Dimbleby Department of Cancer Research, Comprehensive Cancer Centre, Kings College London, London, UK
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Hu
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China; Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, China.
| | - Fubing Wang
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China; Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China; Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China.
| |
Collapse
|
43
|
Wang QY, Xu BY, Wang Y, Lin YM, Zheng LF, Liu G, Li DZ, Jiang CS, Wang W, Zeng XP. Sodium aescinate promotes apoptosis of pancreatic stellate cells and alleviates pancreatic fibrosis by inhibiting the PI3K/Akt/FOXO1 signaling pathways. Front Pharmacol 2025; 16:1554260. [PMID: 40331192 PMCID: PMC12052937 DOI: 10.3389/fphar.2025.1554260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Chronic pancreatitis (CP) is an inflammatory disease of progressive pancreatic fibrosis, and pancreatic stellate cells (PSCs) are key cells involved in pancreatic fibrosis. To date, there are no clinical therapies available to reverse inflammatory damage or pancreatic fibrosis associated with CP. Sodium Aescinate (SA) is a natural mixture of triterpene saponins extracted from the dried and ripe fruits of horse chestnut tree. It has been shown to have anti-inflammatory and anti-edematous effects. This study aims to explore the therapeutic potential of SA in CP and the molecular mechanism of its modulation. Through in vivo animal models and experiments, we found that SA significantly alleviated pancreatic inflammation and fibrosis in caerulein-induced CP mice model. In addition, SA inhibited the proliferation, migration and activation of PSCs as well as promoted apoptosis of PSCs through a series of experiments on cells in vitro including CCK-8 assay, Western blotting, immunofluorescence staining, wound-healing assay, Transwell migration assays, flow cytometric analysis, etc. Further RNA sequencing and in vitro validation assays revealed that inhibition of the PI3K/AKT/FOXO1 signaling pathway was involved in the SA mediated promotion of PSCs apoptosis, thus alleviating pancreatic fibrosis. In conclusion, this study revealed that SA may have promising potential as therapeutic agent for the treatment of CP, and the PI3K/AKT/FOXO1 pathway is a potential therapeutic target for pancreatic inflammation and fibrosis.
Collapse
Affiliation(s)
- Qing-Yun Wang
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Bai-Yan Xu
- Department of Digestive Diseases, Huian County Hospital, Quanzhou, China
| | - Yi Wang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan-Mei Lin
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Lin-Fu Zheng
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Gang Liu
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Da-Zhou Li
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Chuan-Shen Jiang
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wen Wang
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Xiang-Peng Zeng
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| |
Collapse
|
44
|
Bal T. Scaffold-free endocrine tissue engineering: role of islet organization and implications in type 1 diabetes. BMC Endocr Disord 2025; 25:107. [PMID: 40259265 PMCID: PMC12010671 DOI: 10.1186/s12902-025-01919-y] [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: 08/09/2024] [Accepted: 01/17/2025] [Indexed: 04/23/2025] Open
Abstract
Type 1 diabetes (T1D) is a chronic hyperglycemia disorder emerging from beta-cell (insulin secreting cells of the pancreas) targeted autoimmunity. As the blood glucose levels significantly increase and the insulin secretion is gradually lost, the entire body suffers from the complications. Although various advances in the insulin analogs, blood glucose monitoring and insulin application practices have been achieved in the last few decades, a cure for the disease is not obtained. Alternatively, pancreas/islet transplantation is an attractive therapeutic approach based on the patient prognosis, yet this treatment is also limited mainly by donor shortage, life-long use of immunosuppressive drugs and risk of disease transmission. In research and clinics, such drawbacks are addressed by the endocrine tissue engineering of the pancreas. One arm of this engineering is scaffold-free models which often utilize highly developed cell-cell junctions, soluble factors and 3D arrangement of islets with the cellular heterogeneity to prepare the transplant formulations. In this review, taking T1D as a model autoimmune disease, techniques to produce so-called pseudoislets and their applications are studied in detail with the aim of understanding the role of mimicry and pointing out the promising efforts which can be translated from benchside to bedside to achieve exogenous insulin-free patient treatment. Likewise, these developments in the pseudoislet formation are tools for the research to elucidate underlying mechanisms in pancreas (patho)biology, as platforms to screen drugs and to introduce immunoisolation barrier-based hybrid strategies.
Collapse
Affiliation(s)
- Tugba Bal
- Department of Bioengineering, Faculty of Engineering and Natural Sciences, Uskudar University, Istanbul, 34662, Turkey.
| |
Collapse
|
45
|
Wang S, Chen X, Wang K, Yang S. The Regulatory Role of NcRNAs in Pyroptosis and Disease Pathogenesis. Cell Biochem Biophys 2025:10.1007/s12013-025-01720-7. [PMID: 40249522 DOI: 10.1007/s12013-025-01720-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2025] [Indexed: 04/19/2025]
Abstract
Non-coding RNAs (ncRNAs), as critical regulators of gene expression, play a pivotal role in the modulation of pyroptosis and exhibit a close association with a wide range of diseases. Pyroptosis is a form of programmed cell death mediated by inflammasomes, characterized by cell membrane perforation, release of inflammatory cytokines, and a robust immune response. Recent studies have revealed that ncRNAs influence the initiation and execution of pyroptosis by regulating the expression of pyroptosis-related genes or modulating associated signaling pathways. This review systematically summarizes the molecular mechanisms and applications of ncRNAs in diseases such as cancer, infectious diseases, neurological disorders, cardiovascular diseases, and metabolic disorders. It further explores the potential of ncRNAs as diagnostic biomarkers and therapeutic targets, elucidates the intricate interactions among ncRNAs, pyroptosis, and diseases, and provides novel strategies and directions for the precision treatment of related diseases.
Collapse
Affiliation(s)
- Shaocong Wang
- Department of Cardiovascular Surgery, Institute of Chronic Diseases, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Xinzhe Chen
- Department of Cardiovascular Surgery, Institute of Chronic Diseases, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Kun Wang
- Department of Cardiovascular Surgery, Institute of Chronic Diseases, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China.
| | - Sumin Yang
- Department of Cardiovascular Surgery, Institute of Chronic Diseases, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
46
|
Cho SY, Eun HS, Kim J, Ko YD, Rou WS, Joo JS. The Solute Carrier Superfamily as Therapeutic Targets in Pancreatic Ductal Adenocarcinoma. Genes (Basel) 2025; 16:463. [PMID: 40282424 PMCID: PMC12027052 DOI: 10.3390/genes16040463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC), a challenging and malignant cancer, primarily originates from the exocrine cells of the pancreas. The superfamily of solute carrier (SLC) transporters, consisting of more than 450 proteins divided into 65 families, is integral to various cellular processes and represents a promising target in precision oncology. As therapeutic targets, SLC transporters are explored through an integrative analysis. MATERIALS AND METHODS The expression profiles of SLCs were systematically analyzed using mRNA data from The Cancer Genome Atlas (TCGA) and protein data from the Human Protein Atlas (HPA). Survival analysis was examined to evaluate the prognostic significance of SLC transporters for overall survival (OS) and disease-specific survival (DSS). Genetic alterations were examined using cBioPortal, while structural studies were performed with AlphaFold and AlphaMissense to predict functional impacts. Furthermore, Gene Set Enrichment Analysis (GSEA) was carried out to identify oncogenic pathways linked to SLC transporter expression. RESULTS SLC transporters were significantly upregulated in tumors relative to normal tissues. Higher expression levels of SLC39A10 (HR = 1.89, p = 0.0026), SLC22B5 (HR = 1.84, p = 0.0042), SLC55A2 (HR = 2.15, p = 0.00023), and SLC30A6 (HR = 1.90, p = 0.003) were strongly associated with unfavorable OS, highlighting their connection to poor prognosis in PDAC. GSEA highlighted that these four transporters are significantly involved in key oncogenic pathways, such as epithelial-mesenchymal transition (EMT), TNF-α signaling, and angiogenesis. CONCLUSIONS The study identifies four SLCs as therapeutic targets in PDAC, highlighting their crucial role in essential metabolic pathways. These findings lay the groundwork for developing next-generation metabolic anti-cancer treatment to improve survival for PDAC patients.
Collapse
Affiliation(s)
- Sang Yeon Cho
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea;
- CHOMEDICINE Inc., TIPS Town, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Hyuk Soo Eun
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon 35015, Republic of Korea
| | - Jaejeung Kim
- Department of Computer Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea;
| | - Yun Dam Ko
- Seoul Teunteun Rehabilitation Clinic, Jeungpyeong-gun, Chungcheongbuk-do 27937, Republic of Korea
| | - Woo Sun Rou
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
- Department of Internal Medicine, Chungnam National University Sejong Hospital, Sejong 30099, Republic of Korea
| | - Jong Seok Joo
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
- Department of Internal Medicine, Chungnam National University Sejong Hospital, Sejong 30099, Republic of Korea
| |
Collapse
|
47
|
FENG LIWEN, CHEN YUTING, JIN WENYI. Research progress on cancer-associated fibroblasts in osteosarcoma. Oncol Res 2025; 33:1091-1103. [PMID: 40296919 PMCID: PMC12033999 DOI: 10.32604/or.2024.054207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/26/2024] [Indexed: 04/30/2025] Open
Abstract
Osteosarcoma (OS) is a prevalent primary bone malignancy with limited treatment options. Therefore, it is imperative to investigate and understand the mechanisms underlying OS pathogenesis. Cancer-associated fibroblasts (CAFs) are markedly abundant in tumor stromal cells and are essentially involved in the modulation of tumor occurrence and development. In recent years, CAFs have become a hotspot as researchers aim to elucidate CAF mechanisms that regulate tumor progression. However, most studies on CAFs are limited to a few common cancers, and their association with OS remains elusive. This review describes the role and current knowledge of CAFs in OS, focusing on their potential cellular origin, classification, and diverse functionality. It was found that CAFs influenced OS tumor cell signaling, proliferation, invasion, metastasis, epithelial-mesenchymal transition, stemness maintenance, angiogenesis, and the ability to modify immune system components. Furthermore, findings on other common cancers indicated that effective therapeutic strategies included the manipulation of CAF activation, targeting CAF-derived components, and depletion of CAFs by biomarkers. This review provides new insights and a theoretical basis for OS research.
Collapse
Affiliation(s)
- LIWEN FENG
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - YUTING CHEN
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - WENYI JIN
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430022, China
| |
Collapse
|
48
|
NAKASHIMA MOEKA, SUGA NAOKO, FUKUMOTO AKARI, YOSHIKAWA SAYURI, MATSUDA SATORU. Promising roles of vitamin D receptor and APRO family proteins for the development of cancer stem cells targeted malignant tumor therapy. Oncol Res 2025; 33:1007-1017. [PMID: 40296902 PMCID: PMC12034005 DOI: 10.32604/or.2025.059657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/13/2025] [Indexed: 04/30/2025] Open
Abstract
Malignant tumors are heterogeneous diseases characterized by uncontrolled cell proliferation, invasion, metastasis, and/or recurrence of their malignancies. In particular, cancer stem cells (CSCs) within these tumors might be responsible for the property of invasiveness and/or therapies-resistance. CSCs are a self-renewing, awfully tumorigenic subpopulation of cancer cells, which are notorious for strong chemoresistance and are frequently responsible the aggravated invasion, metastasis, and/or recurrence. Developing targeting therapies against CSCs, therefore, may be deliberated a more encouraging mission for the greater cancer therapy. Innovation for a more potent anti-CSC treatment has been required as soon as possible. Interestingly, vitamin D could modulate the inflammatory condition of the tumor microenvironment (TME) by successfully affecting CSCs, which has an imperative role in determining the malignant phenotype of CSCs. In addition, vitamin D may also contribute to the regulation of the malignant behaviors of CSCs. Consistently, vitamin D could have potential applications for the significant inhibition of several tumor growths within various cancer therapies. The biological significance of vitamin D for CSCs regulation may be involved in the function of APRO family proteins. Therefore, vitamin D could be one of the innovative therapeutic modalities for the development of novel CSCs related tumor therapies.
Collapse
Affiliation(s)
- MOEKA NAKASHIMA
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| | - NAOKO SUGA
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| | - AKARI FUKUMOTO
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| | - SAYURI YOSHIKAWA
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| | - SATORU MATSUDA
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| |
Collapse
|
49
|
Nordback I, Paajanen H, Pandol S. How Alcohol Induces Human Acute Alcoholic Pancreatitis-Problem Solved? THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00115-4. [PMID: 40254129 DOI: 10.1016/j.ajpath.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025]
Abstract
It has been a puzzle why only a minority of heavy alcohol drinkers develop acute alcoholic pancreatitis. In this review, the sparse data available from published studies were collected and, based on them, a hypothesis was formed. Long-term high alcohol consumption results in lowered cholecystokinin and cholinergic stimulus of the pancreas, and causes concentration and acidification of pancreatic fluid, predisposing to protein secretion. Early during the withdrawal period when returning to a normal or high-fat nonalcoholic diet, there is a relative hyperstimulation of the pancreas, a well-established mechanism that results in experimental acute pancreatitis. Lower, physiological stimulation is enough to start acute pancreatitis, when the secretions cause temporary obstruction in the duct system; the stimulation against temporary obstruction is also well-known to result in experimental acute pancreatitis. The magnitude of alcohol-induced deficits in acinar cell defense mechanisms then finally determines the onset of pancreatitis.
Collapse
Affiliation(s)
- Isto Nordback
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
| | - Hannu Paajanen
- Department of Gastrointestinal Surgery, University of Eastern Finland, Kuopio, Finland
| | - Stephen Pandol
- Basic and Translational Pancreas Research, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
50
|
Siea AC, Collatuzzo G, Hamdani M, Boffetta P. Welding Fumes Exposure and the Risk of Head and Neck and Gastrointestinal Cancer: A Systematic Review and Meta-Analysis. LA MEDICINA DEL LAVORO 2025; 116:16092. [PMID: 40243545 DOI: 10.23749/mdl.v116i2.16092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 02/14/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND The association between welding fumes and cancers other than lung cancer remains undefined. We conducted a systematic review and meta-analysis on occupational exposure to welding fumes and the risk of head and neck cancer (HN, comprising oral, pharynx, and larynx) and gastrointestinal cancer (GI, comprising esophagus, stomach, colorectal, liver, and pancreas). METHODS A systematic search was performed in PubMed, Scopus, and Embase using PRISMA guidelines. Cohort studies on occupational exposure to welding fumes were identified. Study quality was assessed through the CASP score. Data were analyzed in random-effects models to calculate the relative risks (RR) and 95% confidence intervals (CI) of HN and GI cancer overall and stratified by cancer site. RESULTS Seven independent studies with data on oral, pharynx, larynx, esophagus, stomach, colorectal, liver, or pancreas cancer were identified. We observed the following associations: HN RR=1.10 (95% CI 1.00-1.22); GI RR= 1.03 (95% CI 0.97-1.10); oral and pharynx RR=1.06 (95%, CI 0.93-1.20, eleven risk estimates); larynx RR=1.17 (95%, CI 1.01-1.37, nine risk estimates); esophagus RR=0.98 (95%, CI 0.83-1.15, three risk estimates); stomach RR= 1.10 (95%, CI 1.02-1.19, five risk estimates); colorectal RR=0.99 (95%, CI 0.85-1.15, seven risk estimates); liver RR=1.23 (95%, CI 0.79-1.90, five risk estimates); and pancreas cancer RR=1.05 (95%, CI 0.94-1.16, three risk estimates). CONCLUSIONS We observed an association between occupational exposure to welding fumes and larynx and stomach cancer. No association was found for other HN or GI cancers. Our study stresses the need to investigate the risk of cancers other than lung following occupational exposure to welding fumes.
Collapse
Affiliation(s)
- Andrei Cosmin Siea
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Giulia Collatuzzo
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Maha Hamdani
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|