1
|
Huang X, Wen Z, Cai H, Yu D. The role of quercetin in modulating lipid metabolism and enhancing chemotherapy via the STAT3-CPT1B pathway in pancreatic cancer. Biochem Biophys Res Commun 2025; 772:152033. [PMID: 40412371 DOI: 10.1016/j.bbrc.2025.152033] [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: 02/07/2025] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Pancreatic cancer is a highly aggressive gastrointestinal tumor with limited treatment options, such as surgery and chemotherapy. Thus, further research into its pathogenesis and new treatments is necessary. METHODS Fluorescence-activated cell sorting was employed to sort pancreatic cancer stem cells (PCSCs). Sphere formation assays and Cell Counting Kit-8 (CCK-8) assays were conducted to assess stemness and proliferation capacity. Quantitative real-time PCR and Western blot analysis were employed to assess gene expression levels. Furthermore, immunofluorescence microscopy and chromatin immunoprecipitation assays were conducted to examine alterations in signaling pathways and gene expression. RESULTS Quercetin and gemcitabine may inhibit PANC-1 cells and PCSCs by affecting energy metabolism. Chromatin immunoprecipitation assays revealed an interaction between STAT3 and CPT1B in PCSCs. Quercetin and gemcitabine might affect energy metabolism by inhibiting STAT3 and CPT1B. Manipulating STAT3 expression (overexpression plasmids and siRNA knockdown) altered CPT1B mRNA and protein expression. Although acetyl-CoA reversed the quercetin- and gemcitabine-induced expression of N-cadherin, DECR1, and ALDH, it had minimal influence on CPT1B and STAT3 levels. CONCLUSION Quercetin inhibits the expression of CPT1B via the STAT3 signaling pathway, affecting lipid metabolism and exerting antitumor effects. Furthermore, the combined administration of quercetin and gemcitabine exhibits enhanced therapeutic efficacy.
Collapse
Affiliation(s)
- Xinshi Huang
- Department of Ultrasound, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325003, PR China
| | - Zhengde Wen
- Departments of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325003, PR China
| | - Huajie Cai
- Departments of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325003, PR China
| | - Dinglai Yu
- Departments of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325003, PR China.
| |
Collapse
|
2
|
Bu J, Miao Z, Yang Q. GOT2: New therapeutic target in pancreatic cancer. Genes Dis 2025; 12:101370. [PMID: 40247913 PMCID: PMC12005923 DOI: 10.1016/j.gendis.2024.101370] [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: 11/23/2023] [Revised: 04/18/2024] [Accepted: 06/21/2024] [Indexed: 04/19/2025] Open
Abstract
In recent years, the incidence and mortality rates of pancreatic cancer have been steadily increasing, and conventional therapies have shown a high degree of tolerance. Therefore, the search for new therapeutic targets remains a key issue in current research. Mitochondrial glutamic-oxaloacetic transaminase 2 (GOT2) is an important component of the malate-aspartate shuttle system, which plays an important role in the maintenance of cellular redox balance and amino acid metabolism, and has the potential to become a promising target for anti-cancer therapy. In this paper, we will elaborate on the metabolic and immune effects of GOT2 in pancreatic cancer based on existing studies, with a view to opening up new avenues for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Jiarui Bu
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Zeyu Miao
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Qing Yang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
3
|
Hayashi S, Matsumoto M, Liu L, Tanaka M, Unno M, Igarashi K. Transcription factor BACH1 promotes epithelial-mesenchymal transition by repressing iron metabolism-related genes. Biochem Biophys Res Commun 2025; 767:151898. [PMID: 40315568 DOI: 10.1016/j.bbrc.2025.151898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/28/2025] [Accepted: 04/24/2025] [Indexed: 05/04/2025]
Abstract
Pancreatic adenocarcinoma (PDAC) is one of the cancers with a very poor prognosis for its highly invasive and metastatic ability. Epithelial-mesenchymal transition (EMT) is critical for metastasis and invasion of PDAC cells, in which the expression of epithelial genes, such as E-cadherin (CDH1), decreases and that of mesenchymal genes increases. Transcription factor BTB and CNC homology 1 (BACH1) promotes EMT of PDAC cells in part by indirectly suppressing the expression of CDH1. However, the mechanism behind this is not yet clear. Considering recent reports on a link between intracellular iron and CDH1 expression in cancer cells, we examined whether BACH1 represses CDH1 expression by regulating ferritin gene. When AsPC-1 PDAC cells were treated with the iron chelator deferasirox (DFX), CDH1 expression was increased. While BACH1 knockdown resulted in increased CDH1 expression, combined knockdown of BACH1 and ferritin heavy chain gene (FTH1) reversed CDH1 expression. Tank-binding kinase 1 (TBK1), an upstream regulator of BACH1, was necessary to maintain EMT gene expression patterns in AsPC-1 cells. TBK1 was redundant with BACH1 to maintain VIM expression in SW1990 PDAC cells, suggesting its BACH1-independent role in EMT. Therefore, the regulation of CDH1 and EMT by BACH1 involves FTH1 and intracellular iron as mediators and TBK1 as an upstream and parallel regulator.
Collapse
Affiliation(s)
- Shuichiro Hayashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai, 980-8575, Japan; Department of Surgery, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai, 980-8575, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai, 980-8575, Japan; Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira 2-1-1, Sendai, 980-8577, Japan.
| | - Liang Liu
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai, 980-8575, Japan; Developmental Therapeutics Branch, Center for Cancer Research, NCI Bethesda, MD 20892-4264, USA
| | - Miho Tanaka
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai, 980-8575, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai, 980-8575, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Sendai, 980-8575, Japan.
| |
Collapse
|
4
|
Imaoka H, Ikeda M, Ozaka M, Oshima K, Okano N, Shimizu S, Tsumura H, Komatsu Y, Yamashita T, Kataoka S, Nagano H, Hisano T, Sasaki M, Kobayashi S, Fukushima T, Mitsunaga S, Furukawa T, Hamauchi S, Ueno M, Furuse J. Phase 1/2 study of liposomal irinotecan plus S-1 for metastatic pancreatic cancer refractory to gemcitabine-based treatment. Eur J Cancer 2025; 222:115424. [PMID: 40252631 DOI: 10.1016/j.ejca.2025.115424] [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: 02/05/2025] [Revised: 04/03/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Liposomal irinotecan (nal-IRI) plus fluorouracil/folinic acid (5-FU/LV) improves survival in gemcitabine-refractory metastatic pancreatic cancer (PC) but requires a central venous port. S-1, an oral fluoropyrimidine with proven efficacy in PC, may replace 5-FU/LV in nal-IRI plus 5-FU/LV, potentially enhancing both convenience and antitumor effect. METHODS This single-arm, open-label, phase 1/2 study included patients with histologically or cytologically confirmed adenocarcinoma, aged 20-80 years, an Eastern Cooperative Oncology Group performance status of 0-1, with metastatic disease, and refractory to gemcitabine-based treatment. The primary endpoint in phase 1 part was the frequency of dose-limiting toxicity (DLT) to nal-IRI plus S-1. The primary endpoint in phase 2 part was overall survival. This trial was registered in the Japan Registry of Clinical Trials database (jRCTs031210040). RESULTS In phase 1 part, one patient with DLT was observed at nal-IRI 70 mg/m2 (day 1) with S-1 80 mg/m2/day (day 1-7) in a 2-week cycle, establishing this as the recommended phase 2 dose (RP2D). Forty-nine patients from phase 1 (n = 6) and phase 2 part (n = 43) were treated with the RP2D, and their results were pooled. Median overall survival was 10.3 months (95 % confidence interval, 8.1-12.0 months). A confirmed partial response was achieved in 10 patients (20.4 %). The most frequent treatment-emergent adverse events were hypoalbuminemia (98.0 %), anemia (98.0 %), and anorexia (81.6 %). There were no treatment-related deaths. CONCLUSIONS This study demonstrated that nal-IRI plus S-1 exhibited promising efficacy and an acceptable safety profile in patients with metastatic PC refractory to gemcitabine-based treatment.
Collapse
Affiliation(s)
- Hiroshi Imaoka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kotoe Oshima
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Satoshi Shimizu
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Hidetaka Tsumura
- Department of Gastroenterological Oncology, Hyogo Cancer Center, Akashi, Japan
| | - Yoshito Komatsu
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Taro Yamashita
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa, Japan
| | - Shigeki Kataoka
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Terumasa Hisano
- Department of Hepato-Biliary-Pancreatology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Mitsuhito Sasaki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Satoshi Kobayashi
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Taito Fukushima
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Shuichi Mitsunaga
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takaaki Furukawa
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Satoshi Hamauchi
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Junji Furuse
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|
5
|
Ji W, Xiong Y, Yang W, Shao Z, Guo X, Jin G, Su J, Zhou M. Transcriptomic profiling of blood platelets identifies a diagnostic signature for pancreatic cancer. Br J Cancer 2025; 132:937-946. [PMID: 40133510 DOI: 10.1038/s41416-025-02980-z] [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: 08/02/2024] [Revised: 02/26/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PaCa) is a deadly malignancy that is often diagnosed at an advanced stage, limiting treatment and reducing survival. There is an urgent need for convenient and accurate diagnostic markers for the early detection of PaCa. METHODS In this multicenter case-control study, we performed transcriptome analysis of 673 platelet samples from different in-house and public cohorts. RNA sequencing and RT-qPCR were used to discover and validate potential platelet biomarkers. A multi-gene signature was developed using binomial generalized linear model and independently validated in multicenter cohorts. RESULTS Two platelet RNAs, SCN1B and MAGOHB, consistently showed robust altered expression patterns between PaCa and healthy controls across cohorts, as confirmed by both RNA sequencing and RT-qPCR. The diagnostic two-RNA signature, PLA2Sig, demonstrated remarkable performance in detecting PaCa, with area under the receiver operating characteristic curve (AUC) values of 0.808, 0.900, 0.783, and 0.830 across multicenter cohorts. Furthermore, PLA2Sig effectively identified resectable stage I&II PaCa cases with an AUC of 0.812. Notably, PLA2Sig outperformed the traditional serum markers carcinoembryonic antigen and carbohydrate antigen 19-9 in distinguishing PaCa from healthy controls, and is complementary to established blood-based screening biomarkers. CONCLUSION These findings provide preliminary but promising evidence for the potential utility of platelet RNAs as an alternative non-invasive liquid biopsy tool for the early detection of PaCa.
Collapse
Affiliation(s)
- Weiping Ji
- Department of General Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yichun Xiong
- Department of General Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Wei Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Zhuo Shao
- Department of General Surgery, Shanghai Changhai Hospital of Navy Medical University, Shanghai, 200438, China
| | - Xiaoling Guo
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Gang Jin
- Department of General Surgery, Shanghai Changhai Hospital of Navy Medical University, Shanghai, 200438, China
| | - Jianzhong Su
- Department of General Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| | - Meng Zhou
- Department of General Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
6
|
Crecca E, Di Giuseppe G, Camplone C, Vigiano Benedetti V, Melaiu O, Mezza T, Cencioni C, Spallotta F. The multifaceted role of agents counteracting metabolic syndrome: A new hope for gastrointestinal cancer therapy. Pharmacol Ther 2025; 270:108847. [PMID: 40216262 DOI: 10.1016/j.pharmthera.2025.108847] [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: 10/04/2024] [Revised: 01/27/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
Metabolic syndrome (MetS) is defined by the presence of at least three of five clinical parameters including abdominal obesity, insulin resistance, elevated triglycerides, reduced high-density lipoprotein (HDL) and hypertension. Major features describing MetS have been recognized risk factors for cancer onset, with an alarming impact on gastrointestinal (GI) tumors. Intriguingly, therapeutic administration of drugs to improve glycemic control and dyslipidemia (including metformin, statins) has been shown to have a preventive role in the development and in prognosis improvement of several cancer types. Overall, these observations highlight the key role of altered metabolism prevalently in cancer risk development and unveil anti-MetS agent repurposing potential beyond their conventional pharmacological action. The objective of this review is to summarize the current knowledge about the antitumor activity of anti-diabetic and anti-lipemic agents in GI cancer onset and progression. Here, pre-clinical evidence of their therapeutic potential and of their integration in novel compelling therapeutic strategies will be discussed. Possible clinical outcomes of these novel therapeutic combined protocols specifically dedicated to GI cancer patients will be put under the spotlight. In the future, these novel therapeutic options should be considered to improve conventional chemotherapy response and prognosis of this group of patients.
Collapse
Affiliation(s)
- Elena Crecca
- Institute of System Analysis and Informatics "Antonio Ruberti", National Research Council (IASI-CNR), 00185 Rome, Italy
| | - Gianfranco Di Giuseppe
- Endocrinology and Diabetology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy; Department of Translational Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Claudia Camplone
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185 Rome, Italy; Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | | | - Ombretta Melaiu
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Teresa Mezza
- Department of Translational Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy; Pancreas Unit, CEMAD Digestive Diseases Center, Internal Medicine and Gastroenterology Unit, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Chiara Cencioni
- Institute of System Analysis and Informatics "Antonio Ruberti", National Research Council (IASI-CNR), 00185 Rome, Italy.
| | - Francesco Spallotta
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185 Rome, Italy; Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy.
| |
Collapse
|
7
|
Ramer R, Hinz B. Effect of cannabinoids on the efficacy and side effects of anticancer therapeutic strategies - Current status of preclinical and clinical research. Pharmacol Ther 2025; 270:108851. [PMID: 40221102 DOI: 10.1016/j.pharmthera.2025.108851] [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: 01/31/2025] [Revised: 03/14/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025]
Abstract
Cannabinoids have attracted increasing attention in cancer research in recent decades. A major focus of current preclinical and clinical studies is on the interactions and potential risks when combined with chemotherapeutic agents, targeted therapies and other anticancer strategies. Given the extensive preclinical data on additive, synergistic and, in some cases, antagonistic tumor cell killing effects of chemotherapeutic agents and cannabinoids when co-administered, a critical analysis of these data seems essential. The available data mainly relate to combination treatments for glioblastoma, hematological malignancies and breast cancer, but also for other cancer types. Such an analysis also appears necessary because cannabinoids are used as an option to treat nausea and vomiting caused by chemotherapy, as well as tumor-related pain, and cancer patients sometimes take cannabinoids without a medical prescription. In addition, numerous recent preclinical studies also suggest cannabinoid-mediated relief of other chemotherapy-related side effects such as peripheral neuropathy, nephrotoxicity, cardiotoxicity, cystitis, bladder complications and mucositis. To summarize, the data available to date raise the prospect that cannabinoids may increase the efficacy of chemotherapeutic agents while reducing their side effects. However, preclinical studies on anticancer interactions are mostly limited to cytotoxicity analyses. An equally thorough investigation of the effects of such combinations on the immune system and on the tumorigenic levels of angiogenesis, invasion and metastasis is still pending. On this basis, a comprehensive understanding for the evaluation of a targeted additional treatment of various cancers with cannabinoids could be established.
Collapse
Affiliation(s)
- Robert Ramer
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany
| | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany.
| |
Collapse
|
8
|
Hou Y, Lv B, Du J, Ye M, Jin H, Yi Y, Huang Y. Sulfide regulation and catabolism in health and disease. Signal Transduct Target Ther 2025; 10:174. [PMID: 40442106 PMCID: PMC12122839 DOI: 10.1038/s41392-025-02231-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/03/2025] [Accepted: 03/21/2025] [Indexed: 06/02/2025] Open
Abstract
The metabolic pathway of sulfur-containing amino acids in organisms begins with methionine, which is metabolized to produce important sulfur-containing biomolecules such as adenosylmethionine, adenosylhomocysteine, homocysteine, cystine, and hydrogen sulfide (H2S). These sulfur-containing biomolecules play a wide range of physiological roles in the body, including anti-inflammation, antioxidant stress, DNA methylation, protein synthesis, etc., which are essential for maintaining cellular function and overall health. In contrast, dysregulation of the metabolic pathway of sulfur-containing amino acids leads to abnormal levels of sulfur-containing biomolecules, which produce a range of pathological consequences in multiple systems of the body, such as neurodegenerative diseases, cardiovascular diseases, and cancer. This review traces the milestones in the development of these sulfur-containing biomolecules from their initial discovery to their clinical applications and describes in detail the structure, physiochemical properties, metabolism, sulfide signaling pathway, physiopathological functions, and assays of sulfur-containing biomolecules. In addition, the paper also explores the regulatory role and mechanism of sulfur-containing biomolecules on cardiovascular diseases, liver diseases, neurological diseases, metabolic diseases and tumors. The focus is placed on donors of sulfur-containing biological macromolecule metabolites, small-molecule drug screening targeting H2S-producing enzymes, and the latest advancements in preclinical and clinical research related to hydrogen sulfide, including clinical trials and FDA-approved drugs. Additionally, an overview of future research directions in this field is provided. The aim is to enhance the understanding of the complex physiological and pathological roles of sulfur-containing biomolecules and to offer insights into developing effective therapeutic strategies for diseases associated with dysregulated sulfur-containing amino acid metabolism.
Collapse
Affiliation(s)
- Yuanyuan Hou
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, Beijing, 100034, China
| | - Boyang Lv
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, Beijing, 100034, China
| | - Junbao Du
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, Beijing, 100034, China
| | - Min Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Yunnan Baiyao International Medical Research Center, Peking University, 38 Xueyuan Road, Beijing, 100191, China
| | - Hongfang Jin
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, Beijing, 100034, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
| | - Yang Yi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- Yunnan Baiyao International Medical Research Center, Peking University, 38 Xueyuan Road, Beijing, 100191, China.
| | - Yaqian Huang
- Department of Pediatrics, Children's Medical Center, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
9
|
Selvam SP, Kamalakannan S, Mathivanan A, Cho S. Engineering Pt Single-Atom Doped SeS 2/Ti 3CNT x MXene with Molecularly Imprinted Polymer for Precision Pancreatic Cancer Diagnostics: DFT and Molecular Dynamics Perspectives. SMALL METHODS 2025:e2500475. [PMID: 40420649 DOI: 10.1002/smtd.202500475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 05/11/2025] [Indexed: 05/28/2025]
Abstract
Cyclophilin-B (CypB) is overexpressed in pancreatic cancer, thus, the potential screening of CypB in biofluids and tissue samples may boost the identification of early-stage pancreatic cancer. A novel strategy of CypB detection utilizing the molecularly imprinted polymer platform, comprising higher binding affinity exhibiting cavities against the CypB protein was developed. Specifically, a nanocatalyst consisting of Pt single atom (Ptsa)-doped selenium disulfide (SeS2)/Ti3CNTx MXene nanocomposite is designed. The sluggish diffusion of Ptsa caused by the highest migration energy barrier of 6.39 eV unveils exceptionally high stability (2.89 ×1088 d (300 K) and 1.053 × 1024 d (750 K)) with (SeS2)/Ti3CNTx surface. The Ptsa boosted charge transfer kinetics paves the improved performance of the CypB sensor, while SeS2/Ti3CNTx supports the stable current density overall. The system establishes a dynamic linear range from 0.12 to 250 nm of CypB detection which correlates with the physiological existence of the CypB in human biofluids and tissues and the excellent detection limit of 80 pm. The liquid chromatography integrated mass spectrometer investigation warranted the significant enhancement of CypB associates with the progression of pancreatic cancer.
Collapse
Affiliation(s)
- Sathish Panneer Selvam
- Department of Electronic Engineering, Gachon University, Seongnam-si, Gyeonggi-do, 13120, South Korea
| | | | - Agalya Mathivanan
- Department of Physics, Sri Manakula Vinayagar Engineering College, Madagadipet, Puducherry, 605107, India
| | - Sungbo Cho
- Department of Electronic Engineering, Gachon University, Seongnam-si, Gyeonggi-do, 13120, South Korea
- Department of Health Sciences and Technology, Gachon University, Incheon, 21999, South Korea
- Department of Semiconductor Engineering, Gachon University, Seongnam-si, Gyeonggi-do, 13120, South Korea
| |
Collapse
|
10
|
Lin J, Li Y, Sun J. Modulating immune cells within pancreatic ductal adenocarcinoma via nanomedicine. Essays Biochem 2025:EBC20243001. [PMID: 40420798 DOI: 10.1042/ebc20243001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/28/2025] [Indexed: 05/28/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy characterized by a dense extracellular matrix (ECM) and a uniquely immunosuppressive tumor microenvironment (TME), which together form a formidable barrier that hinders deep drug penetration, limiting the efficacy of conventional therapies and leading to poor patient outcomes. Nanocarrier technology emerges as a promising strategy to improve treatment efficacy in PDAC. Nanocarriers can not only improve drug penetration through their adjustable physicochemical properties but also effectively regulate immune cell function in pancreatic cancer TME and promote anti-tumor immune response. This mini-review discusses the effects of nanocarriers on the immune microenvironment of PDAC, analyzing their mechanisms in modulating immune cells, overcoming ECM barriers, and reshaping the TME.
Collapse
Affiliation(s)
- Junyi Lin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, U.S.A
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68106, U.S.A
| | - Ying Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, U.S.A
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68106, U.S.A
| | - Jingjing Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, U.S.A
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68106, U.S.A
| |
Collapse
|
11
|
He R, Shen Z, Chen Q, Hu H, Ding X, Zheng Z, Feng Q, Li B. Pancreatic cancer mortality in China from 2004 to 2021: an in-depth analysis of age, gender, and regional disparities. BMC Cancer 2025; 25:891. [PMID: 40389880 PMCID: PMC12087137 DOI: 10.1186/s12885-025-13863-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/04/2025] [Indexed: 05/21/2025] Open
Abstract
OBJECTIVE This study aimed to analyze the trends and epidemiological characteristics of pancreatic cancer (PC) mortality in China from 2004 to 2021, focusing on gender, age, and regional disparities. The goal was to provide a comprehensive understanding of PC mortality and identify key risk factors to support future prevention and control strategies. METHODS Using data from the national Disease Surveillance Point (DSP) system, which covers a large and representative sample of the Chinese population, the study examined pancreatic cancer mortality trends across different age groups, sexes, and regions. Statistical analyses, including the independent-sample t-test and age-period-cohort (APC) model, were employed to assess mortality differences and annual percentage changes from 2004 to 2021. RESULTS The study recorded a significant increase in pancreatic cancer mortality, particularly among males and older adults. Mortality was consistently higher in urban areas, but the growth rate in rural areas surpassed that of urban areas. Regional disparities were also observed, with the eastern region showing the highest mortality rates but slower increases compared to the central and western regions. Key risk factors, including aging, diabetes, smoking, and chronic pancreatitis, were identified, with gender-specific differences linked to lifestyle factors such as smoking and alcohol consumption. CONCLUSION Pancreatic cancer mortality in China has shown significant increases over the past 18 years, especially among males, older adults, and rural populations. The findings highlight the urgent need for targeted public health interventions to address gender- and age-specific risks, as well as healthcare access inequalities in less developed regions. Future research should focus on gathering more granular, individual-level data to better understand the complex interplay of risk factors and inform more effective prevention and treatment strategies.
Collapse
Affiliation(s)
- Rui He
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhengnan Shen
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiuping Chen
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Haiyang Hu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xin Ding
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhenglong Zheng
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Quansheng Feng
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Baixue Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
12
|
Hesami Z, Sabzehali F, Khorsand B, Alipour S, Sadeghi A, Asri N, Pazienza V, Houri H. Microbiota as a state-of-the-art approach in precision medicine for pancreatic cancer management: A comprehensive systematic review. iScience 2025; 28:112314. [PMID: 40276756 PMCID: PMC12019022 DOI: 10.1016/j.isci.2025.112314] [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: 10/27/2024] [Revised: 12/22/2024] [Accepted: 03/25/2025] [Indexed: 04/26/2025] Open
Abstract
Emerging evidence suggests that harnessing the microbiome holds promise for innovative diagnostic and therapeutic strategies in the management of pancreatic cancer (PC). This study aims to systematically summarize the microbial markers associated with PC and assess their potential application in clinical outcome. Forty-one studies were included to assess the associations between microbial markers and PC. Among these, 13 were developed prediction models related to the microbiome in which the highest diagnostic and prognostic model belong to blood and intratumor markers, respectively. Notably, findings that utilize microbiotas from various body sites were elucidated, demonstrating their importance as unique signatures in biomarker discovery for diverse clinical applications. This review provides unique perspectives on overcoming challenges in PC by highlighting potential microbial-related markers as non-invasive approaches. Further clinical studies should evaluate the utility and accuracy of key indicators in the microbiome as a personalized tool for managing PC.
Collapse
Affiliation(s)
- Zeinab Hesami
- Student Research Committee, Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fattaneh Sabzehali
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Khorsand
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Samira Alipour
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nastaran Asri
- Celiac Disease and Gluten Related Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Valerio Pazienza
- Division of Gastroenterology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Hamidreza Houri
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Brugiapaglia S, Spagnolo F, Curcio C. Unlocking the Potential of Bioactive Compounds in Pancreatic Cancer Therapy: A Promising Frontier. Biomolecules 2025; 15:725. [PMID: 40427617 PMCID: PMC12109016 DOI: 10.3390/biom15050725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a highly challenging malignancy to treat, with a high mortality rate and limited therapeutic options. Despite advances in cancer research, the prognosis for patients diagnosed with PDA is often poor due to late-stage detection and resistance to conventional therapies. Consequently, there is growing interest in the potential of bioactive compounds as alternative or adjuvant treatments, given their ability to target multiple aspects of cancer biology, offering a more holistic approach to treatment. In the context of PDA, certain bioactive compounds, such as polyphenols (found in fruits, vegetables, and tea), flavonoids, carotenoids and compounds in cruciferous vegetables, have shown potential in inhibiting cancer cell growth, reducing inflammation, and promoting cancer cell apoptosis. This review aims to elucidate the mechanisms, by which these bioactive compounds exert their effects, modulating the oxidative stress, influencing inflammatory pathways and regulating cell survival and death. It also highlights current clinical trials that are paving the way toward incorporating these natural agents into mainstream treatment strategies, with the goal of boosting the efficacy of conventional therapies for PDA.
Collapse
Affiliation(s)
- Silvia Brugiapaglia
- Department of Molecular Biotechnology and Health Sciences, Piazza Nizza 44b, 10126 Turin, Italy;
| | - Ferdinando Spagnolo
- School of Advanced Defence Studies, Defence Research & Analysis Institute, Piazza della Rovere 83, 00165 Rome, Italy;
- Defense Institute for Biomedical Sciences, Via Santo Stefano Rotondo 4, 00184 Rome, Italy
| | - Claudia Curcio
- Department of Molecular Biotechnology and Health Sciences, Piazza Nizza 44b, 10126 Turin, Italy;
| |
Collapse
|
14
|
Zhao S, Gao Z, Hu L, Li Y, Wang X, Li X, Chen M, Chen F, Song Z. Reversing VTN deficiency inhibits the progression of pancreatic cancer and enhances sensitivity to anti-PD1 immunotherapy. Front Immunol 2025; 16:1578870. [PMID: 40433359 PMCID: PMC12106453 DOI: 10.3389/fimmu.2025.1578870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/17/2025] [Indexed: 05/29/2025] Open
Abstract
Background Pancreatic cancer, a highly lethal malignancy with limited therapeutic options, necessitates the identification of novel prognostic biomarkers and therapeutic targets. The extracellular matrix protein vitronectin (VTN) has been implicated in tumor progression, but its specific role in pancreatic cancer progression and immunotherapy response remains unclear. Methods This study employed an integrative approach combining single-cell RNA sequencing, analysis of public databases, and functional assays. In vitro experiments assessed the impact of VTN knockdown and overexpression on pancreatic cancer cell proliferation, invasion, and migration. Mechanistic investigations explored associations between VTN expression and immune regulatory factors. A syngeneic mouse subcutaneous tumor model evaluated the therapeutic efficacy of VTN overexpression combined with anti-PD1 immunotherapy. Results VTN was significantly downregulated in pancreatic cancer tissues compared to normal tissues. Lower VTN levels correlated with poorer overall survival. VTN knockdown promoted pancreatic cancer cell proliferation, invasion, and migration in vitro, whereas VTN overexpression suppressed these phenotypes. VTN expression was linked to immune regulatory pathways. High VTN levels predicted improved survival in patients receiving anti-PD1/PD-L1 therapy. In a mouse model, VTN overexpression inhibited tumor growth and synergized with anti-PD1 therapy to enhance antitumor efficacy, suggesting combinatorial therapeutic potential. Conclusions This study identifies VTN as a dual-functional regulator in pancreatic cancer, acting as both a suppressor of tumor progression and a modulator of immunotherapy response. These findings position VTN as a prognostic biomarker and a therapeutic target to sensitize pancreatic tumors to anti-PD1-based immunotherapy, providing a potential strategy for overcoming treatment resistance in this aggressive malignancy.
Collapse
Affiliation(s)
- Siqi Zhao
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhaofeng Gao
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Lingyu Hu
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Yihan Li
- Department of Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoguang Wang
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiaoping Li
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Minjie Chen
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Fei Chen
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhengwei Song
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
15
|
Papakonstantinou D, Wang H, Bani MA, Mulder K, Dunsmore G, Boilève A, Jules-Clément G, Panunzi L, de Sousa LR, de la Calle Fabregat C, Deloger M, Signolle N, Gessain G, Nikolaev SI, Ducreux M, Hollebecque A, Ginhoux F, Blériot C. Molecular analysis highlights TREM2 as a discriminating biomarker for patients suffering from pancreatic ductal adenocarcinoma. Cancer Treat Res Commun 2025; 43:100939. [PMID: 40354768 DOI: 10.1016/j.ctarc.2025.100939] [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: 05/02/2025] [Accepted: 05/02/2025] [Indexed: 05/14/2025]
Abstract
Pancreatic cancer is projected to become the second leading cause of cancer-related deaths by 2030, with its mortality continuing to rise, unlike other common cancers such as breast or colorectal. Late-stage diagnosis, often accompanied by metastatic dissemination, drastically impairs patient survival and underscores the urgent need for improved biomarkers to guide therapeutic strategies. While molecular signatures have been proposed to stratify pancreatic cancer patients, their ability to predict outcomes remains limited. In this study, we applied established molecular signatures to our in-house transcriptomic data from a cohort of pancreatic cancer patients. We took advantage of published datasets to construct comprehensive atlases of cells present in primary and metastatic pancreatic cancers. The atlas of metastasis samples, representative of routinely harvested patient biopsies, revealed that monocyte/macrophage signatures provided superior discriminatory power compared to existing molecular classifications. Notably, the abundance of TREM2-expressing macrophages emerged as a significant parameter for stratifying patients. Our findings position TREM2+ macrophages as a promising biomarker for pancreatic cancer, with potential to enhance patient stratification and inform the development of targeted therapies. This work highlights the critical role of tumor-associated macrophages in pancreatic cancer progression and lays the groundwork for further functional and translational studies.
Collapse
Affiliation(s)
| | - Haiding Wang
- Gustave Roussy Cancer Campus, CNRS UMR9018, Villejuif, France
| | - Mohamed-Amine Bani
- Gustave Roussy Cancer Campus, Medical oncology department, Villejuif, France
| | - Kevin Mulder
- Gustave Roussy Cancer Campus, INSERM U1015, Villejuif, France
| | - Garett Dunsmore
- Gustave Roussy Cancer Campus, INSERM U1015, Villejuif, France
| | - Alice Boilève
- Gustave Roussy Cancer Campus, INSERM U1279, Université Paris Saclay, Villejuif, France; Gustave Roussy Cancer Campus, Biopathology department, Villejuif, France
| | - Gérôme Jules-Clément
- Gustave Roussy Cancer Campus, Bioinformatics Core Facility, CNRS, INSERM, Université Paris-Saclay, Villejuif, France
| | - Leonardo Panunzi
- Gustave Roussy Cancer Campus, INSERM U981, Université Paris Saclay, Villejuif, France
| | | | | | - Marc Deloger
- Gustave Roussy Cancer Campus, Bioinformatics Core Facility, CNRS, INSERM, Université Paris-Saclay, Villejuif, France
| | - Nicolas Signolle
- Gustave Roussy Cancer Campus, Biopathology department, Villejuif, France
| | - Grégoire Gessain
- Gustave Roussy Cancer Campus, Biopathology department, Villejuif, France
| | - Sergey I Nikolaev
- Gustave Roussy Cancer Campus, INSERM U981, Université Paris Saclay, Villejuif, France
| | - Michel Ducreux
- Gustave Roussy Cancer Campus, Medical oncology department, Villejuif, France
| | - Antoine Hollebecque
- Gustave Roussy Cancer Campus, Medical oncology department, Villejuif, France
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, INSERM U1015, Villejuif, France
| | - Camille Blériot
- Gustave Roussy Cancer Campus, INSERM U1015, Villejuif, France; Gustave Roussy Cancer Campus, CNRS UMR9018, Villejuif, France; Institut Necker Enfants Malades, CNRS, INSERM, Université Paris Cité, Paris, France.
| |
Collapse
|
16
|
Ahn HM, Jung BK, Hong J, Hong D, Yoon AR, Yun CO. Enhanced potency of immune checkpoint inhibitors against poorly immunological solid tumors by immune stimulatory oncolytic adenoviruses-mediated remodeling of the tumor microenvironment. Mol Med 2025; 31:175. [PMID: 40335925 PMCID: PMC12057182 DOI: 10.1186/s10020-025-01223-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/22/2025] [Indexed: 05/09/2025] Open
Abstract
Immune checkpoint inhibitor (ICI) have shown promising results against a variety of solid tumors across clinical trials. However, ICI monotherapy is often ineffective in patients with non-immunogenic tumors that exhibit high level of immunosuppression and low level of tumor infiltrating lymphocytes. To address these limitations, we have investigated a combination of ICIs [anti-PD-1 antibody (αPD-1), anti-PD-L1 antibody (αPD-L1), or anti-CTLA-4 antibody (αCTLA-4)] with several different immune stimulatory oncolytic adenoviruses (Ads) expressing different combinations of antitumor cytokines or immune modulatory factors [e.g., (1) interleukin (IL)-12 and granulocyte-macrophage colony-stimulating factor (GM-CSF; RdB/IL12/GMCSF), (2) IL-12 and short hairpin ribonucleic acid (shRNA) targeting vascular endothelial growth factor (RdB/IL12/shVEGF), (3) IL-12 and decorin (RdB/IL12/DCN), (4) GM-CSF, and thymidine kinase (RdB/IL12/GMCSF-TK), or (5) IL-12, GM-CSF, and relaxin (RdB/IL12/GMCSF-RLX)] to overcome tumor-induced immunosuppression. Through comparative evaluation of combination therapy regimens, our findings have identified αPD-1 as the optimal ICI candidate to synergize with different oncolytic Ads to induce potent antitumor immune response against poorly immunological solid tumors.
Collapse
Affiliation(s)
- Hyo Min Ahn
- GeneMedicine Co., Ltd, 222 Wangsimni-Ro, Seongdong-Gu, Seoul, Republic of Korea
| | - Bo-Kyeong Jung
- GeneMedicine Co., Ltd, 222 Wangsimni-Ro, Seongdong-Gu, Seoul, Republic of Korea
| | - JinWoo Hong
- GeneMedicine Co., Ltd, 222 Wangsimni-Ro, Seongdong-Gu, Seoul, Republic of Korea
| | - Dayoung Hong
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea.
- Institute of Nano Science and Technology (INST), Hanyang University, Seoul, Republic of Korea.
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, Republic of Korea.
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea.
- Institute of Nano Science and Technology (INST), Hanyang University, Seoul, Republic of Korea.
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, Republic of Korea.
- GeneMedicine Co., Ltd, 222 Wangsimni-Ro, Seongdong-Gu, Seoul, Republic of Korea.
| |
Collapse
|
17
|
Li X, Su Y, Lin N, Chen Y, Li Z, Zhang Z, Zhao X, Zeng H. Lycium barbarum Polysaccharide-Stabilized Selenium Nanoparticles Deliver Triptolide to Induce Apoptosis for Pancreatic Cancer In Vitro and In Vivo. ACS OMEGA 2025; 10:17108-17122. [PMID: 40352487 PMCID: PMC12059884 DOI: 10.1021/acsomega.4c04743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 04/06/2025] [Accepted: 04/14/2025] [Indexed: 05/14/2025]
Abstract
Pancreatic cancer (PC), known as the ″king of cancer," is a prevalent and aggressive form of malignant tumor affecting the digestive tract. Triptolide (TP), an epoxidized diterpenoid lactone extracted from Tripterygium wilfordii, shows promising antitumor activity. However, the systemic toxicity and poor water solubility of TP inhibit its clinical application. In this work, Lycium barbarum polysaccharide (LBP)-modified selenium nanoparticles (SeNPs), capable of reducing severe toxicity and enhancing solubility, have been designed, synthesized, and applied for the treatment of PC. In vitro release results revealed that TP showed an acid-dependent and sustained-release effect. In Pan02 cells, the IC50 values of TP and LBP-SeNPs@TP were 26.03 ± 2.82 ng/mL and 11.80 ± 2.64 ng/mL, respectively. Similarly, the IC50 values of TP and LBP-SeNPs@TP for L02 cells were 15.76 ± 0.58 ng/mL and 32.73 ± 2.61 ng/mL, suggesting enhanced antitumor efficacy along with reduced toxicity. Flow cytometry analysis demonstrated that LBP-SeNPs@TP exerted the most potent apoptotic effect, achieving an early apoptosis rate of 24.5% and a late apoptosis rate of 45.3%. Notably, the mitochondrial membrane potential was significantly reduced, while ROS production was increased in the LBP-SeNPs@TP group. LBP-SeNPs@TP could significantly inhibit tumor growth while minimizing toxicity. RT-qPCR analysis demonstrated that LBP-SeNPs@TP upregulated the mRNA expression of Bax, Cyt C, and Caspase-3, while downregulating Bcl-2 expression in vitro and in vivo. The immunohistochemical analysis of the tumor tissue further confirmed these results. Overall, LBP-SeNPs emerge as a promising platform for poorly soluble drugs, offering a potential therapeutic approach for pancreatic cancer.
Collapse
Affiliation(s)
- Xiaofang Li
- Academy
of Chinese Medicine Sciences, Henan University
of Chinese Medicine, Zhengzhou 450046, PR China
- The
First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450099, PR China
| | - Yunfang Su
- Academy
of Chinese Medicine Sciences, Henan University
of Chinese Medicine, Zhengzhou 450046, PR China
- The
First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450099, PR China
| | - Na Lin
- Rehabilitation
Department, Kaifeng Hospital of Traditional
Chinese Medicine, Kaifeng 475000, PR China
| | - Yujie Chen
- Academy
of Chinese Medicine Sciences, Henan University
of Chinese Medicine, Zhengzhou 450046, PR China
| | - Zhonghua Li
- Academy
of Chinese Medicine Sciences, Henan University
of Chinese Medicine, Zhengzhou 450046, PR China
| | - Zhenqiang Zhang
- Academy
of Chinese Medicine Sciences, Henan University
of Chinese Medicine, Zhengzhou 450046, PR China
| | - Xuebin Zhao
- Technology
Center, China Tobacco Henan Industrial Co.,
Ltd., Zhengzhou 450000, PR China
| | - Huahui Zeng
- Academy
of Chinese Medicine Sciences, Henan University
of Chinese Medicine, Zhengzhou 450046, PR China
| |
Collapse
|
18
|
Ge H, Wolters-Eisfeld G, Hackert T, Li Y, Güngör C. Development of a hypoxia-responsive macrophage prognostic model using single-cell and bulk RNA sequencing in pancreatic cancer. PLoS One 2025; 20:e0322618. [PMID: 40315225 PMCID: PMC12047781 DOI: 10.1371/journal.pone.0322618] [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: 08/30/2024] [Accepted: 03/24/2025] [Indexed: 05/04/2025] Open
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma (PDAC) is characterized by a low survival rate and limited responsiveness to current therapies. The role of hypoxia in the tumor microenvironment is critical, influencing tumor progression and therapy resistance. The aim of this study was to implement the complex dynamics of the hypoxic tumor microenvironment in PDAC in a hypoxia-related prognosis model. METHODS We utilized single-cell RNA sequencing (scRNA-seq) data and integrated it with TCGA-PAAD database to identify hypoxia-responsive macrophage subsets and related genes. Kaplan-Meier survival analysis, Cox regression, and Lasso regression methods were employed to construct and validate a hypoxia-related prognostic model. The model's effectiveness was evaluated through its predictive capabilities regarding chemotherapy sensitivity and overall survival. RESULTS Our research integrated data from scRNA-seq and the TCGA-PAAD database to construct a hypoxia-related prognostic model that encompassed 13 critical genes. This hypoxia model independently predicted chemotherapy response and poor outcomes, outperforming traditional clinicopathologic features. Additionally, a pan-cancer analysis affirmed the relevance of our hypoxia-related genes across multiple malignancies, particularly highlighting KRTCAP2 as a pivotal biomarker associated with worse prognosis and reduced immune infiltration. CONCLUSION Our findings underscored the prognostic potential of hypoxia-related model and offered a novel avenue for therapeutic targeting, aiming to ameliorate outcomes in pancreatic cancer.
Collapse
Affiliation(s)
- Heming Ge
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerrit Wolters-Eisfeld
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yuqiang Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Cenap Güngör
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
19
|
Gofrit ON, Gofrit B, Popovtzer A, Sosna J, Goldberg SN. The Clonal Trajectory of Liver and Lung Metastases in Pancreatic Ductal Adenocarcinoma. Cancer Rep (Hoboken) 2025; 8:e70228. [PMID: 40348585 PMCID: PMC12063064 DOI: 10.1002/cnr2.70228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/16/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Metastatic spread can follow either the linear route-dissemination of fully malignant cells from the primary tumor, or the parallel route-dissemination of immature tumor cells and independent maturation to metastases in target organs. The linear/parallel ratio (LPR) is a model that uses metastases diameter comparisons to decipher dissemination route. LPR of +1 suggests pure linear and -1 pure parallel spread. AIMS To examine the metastases trajectory in pancreatic duct adenocarcinoma (PDAC). METHODS AND RESULTS A total of 133 patients with PDAC, including 97 patients (72.9%) with synchronous and 36 (27.1%) with metachronous metastases with a total of 1054 lung and 2898 liver metastases, were evaluated. We found that metastatic spread to both liver and lungs is almost exclusively via the linear route (lungs median LPR + 1, interquartile range [IQR] 0.97,1. Liver median LPR + 0.98, IQR 0.83,1). Calculated from the primary diagnosis, overall survival (OS) of patients with metachronous metastases was significantly better compared to patients with synchronous disease (14 months, IQR 10,26, vs. 7 months, IQR 6,9, p < 0.0001). However, calculated from the time of metastases diagnosis, OS of both groups was similar (4 months, IQR 3,8, vs. 7 months, IQR 6,9, p = 0.235). CONCLUSION These two observations suggest that metastatic spread of PDAC is almost exclusively via the linear route, that is, directly from the primary tumor. Therefore, liver or lung metastases are already present in most patients with PDAC at the time of initial diagnosis. This suggests that local treatment in patients with seemingly localized disease does not decrease their risk of developing metastases and that systemic treatment must follow.
Collapse
Affiliation(s)
- Ofer N. Gofrit
- Department of UrologyHadassah Medical Center, Faculty of Medicine, Hebrew University of JerusalemJerusalemIsrael
| | - Ben Gofrit
- School of Engineering and Computer Science, Hebrew University of JerusalemJerusalemIsrael
| | - Aron Popovtzer
- Department of OncologyHadassah Medical Center, Faculty of Medicine, Hebrew University of JerusalemJerusalemIsrael
| | - Jacob Sosna
- Department of RadiologyHadassah Medical Center, Faculty of Medicine, Hebrew University of JerusalemJerusalemIsrael
| | - S. Nahum Goldberg
- Department of RadiologyHadassah Medical Center, Faculty of Medicine, Hebrew University of JerusalemJerusalemIsrael
| |
Collapse
|
20
|
Yang C, Wu Y, Tu H, Yeh Y, Lin TE, Sung T, Li M, Yen S, Hsieh J, Yu M, Hsieh S, Hsieh H, Pan S, Hsu K. Identification and Biological Evaluation of a Novel CLK4 Inhibitor Targeting Alternative Splicing in Pancreatic Cancer Using Structure-Based Virtual Screening. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416323. [PMID: 40126184 PMCID: PMC12097107 DOI: 10.1002/advs.202416323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/02/2025] [Indexed: 03/25/2025]
Abstract
Pancreatic cancer is an aggressive malignancy with a poor prognosis and limited treatment options. Cdc-like kinase 4 (CLK4), a kinase that regulates alternative splicing by phosphorylating spliceosome components, is implicated in aberrant splicing events driving pancreatic cancer progression. In this study, we established a computational model that integrates pharmacological interactions of CLK4 inhibitors with an improved hit rate. Through this model, we identified a novel CLK4 inhibitor, compound 150441, with a 50% inhibitory concentration (IC50) value of 21.4 nm. Structure-activity relationship analysis was performed to investigate key interactions and functional groups. Kinase profiling revealed that compound 150441 is selective for CLK4. Subsequent in vitro assays demonstrated that this inhibitor effectively suppressed cell growth and viability of pancreatic cancer cells. In addition, it inhibited the phosphorylation of key splicing factors, including serine- and arginine-rich splicing factor (SRSF) 4 and SRSF6. Cell cycle analysis further indicated that the compound induced G2/M arrest, leading to apoptosis. RNA-seq analysis revealed that the compound induced significant changes in alternative splicing and key biological pathways, including RNA processing, DNA replication, DNA damage, and mitosis. These findings suggest that compound 150441 has promising potential for further development as a novel pancreatic cancer treatment.
Collapse
Affiliation(s)
- Chun‐Lin Yang
- Graduate Institute of Cancer Biology and Drug DiscoveryCollege of Medical Science and TechnologyTaipei Medical UniversityTaipei110301Taiwan
| | - Yi‐Wen Wu
- Graduate Institute of Cancer Biology and Drug DiscoveryCollege of Medical Science and TechnologyTaipei Medical UniversityTaipei110301Taiwan
| | - Huang‐Ju Tu
- Graduate Institute of Cancer Biology and Drug DiscoveryCollege of Medical Science and TechnologyTaipei Medical UniversityTaipei110301Taiwan
| | - Yun‐Hsuan Yeh
- Graduate Institute of Cancer Biology and Drug DiscoveryCollege of Medical Science and TechnologyTaipei Medical UniversityTaipei110301Taiwan
| | - Tony Eight Lin
- Graduate Institute of Cancer Biology and Drug DiscoveryCollege of Medical Science and TechnologyTaipei Medical UniversityTaipei110301Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug DiscoveryCollege of Medical Science and TechnologyTaipei Medical UniversityTaipei110301Taiwan
| | - Tzu‐Ying Sung
- Graduate Institute of Cancer Biology and Drug DiscoveryCollege of Medical Science and TechnologyTaipei Medical UniversityTaipei110301Taiwan
| | - Mu‐Chun Li
- Institute of Biotechnology and Pharmaceutical ResearchNational Health Research InstitutesMiaoli County350401Taiwan
- Biomedical Translation Research CenterAcademia SinicaTaipei115202Taiwan
| | - Shih‐Chung Yen
- Warshel Institute for Computational BiologySchool of MedicineThe Chinese University of Hong Kong (Shenzhen)ShenzhenGuangdong518172China
| | - Jui‐Hua Hsieh
- Division of Translational ToxicologyNational Institute of Environmental Health SciencesNational Institutes of HealthDurhamNC27709USA
| | - Ming‐Chin Yu
- College of MedicineChang Gung UniversityTaoyuan333323Taiwan
- Department of SurgeryNew Taipei Municipal TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation)TuchengNew Taipei City236043Taiwan
- Graduate Institute of Clinical Medical SciencesChang Gung UniversityGuishanTaoyuan333323Taiwan
| | - Sen‐Yung Hsieh
- College of MedicineChang Gung UniversityTaoyuan333323Taiwan
- Department of Gastroenterology and HepatologyChang Gung Memorial HospitalLinkouTaoyuan333423Taiwan
| | - Hsing‐Pang Hsieh
- Institute of Biotechnology and Pharmaceutical ResearchNational Health Research InstitutesMiaoli County350401Taiwan
- Biomedical Translation Research CenterAcademia SinicaTaipei115202Taiwan
- Department of ChemistryNational Tsing Hua UniversityHsinchu300044Taiwan
| | - Shiow‐Lin Pan
- Graduate Institute of Cancer Biology and Drug DiscoveryCollege of Medical Science and TechnologyTaipei Medical UniversityTaipei110301Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug DiscoveryCollege of Medical Science and TechnologyTaipei Medical UniversityTaipei110301Taiwan
- TMU Research Center of Cancer Translational MedicineTaipei Medical UniversityTaipei110301Taiwan
| | - Kai‐Cheng Hsu
- Graduate Institute of Cancer Biology and Drug DiscoveryCollege of Medical Science and TechnologyTaipei Medical UniversityTaipei110301Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug DiscoveryCollege of Medical Science and TechnologyTaipei Medical UniversityTaipei110301Taiwan
- TMU Research Center of Cancer Translational MedicineTaipei Medical UniversityTaipei110301Taiwan
- Cancer CenterWan Fang Hospital, Taipei Medical UniversityTaipei116079Taiwan
| |
Collapse
|
21
|
Lin Q, Guan S, Peng M, Zhang K, Zhang H, Mo T, Yu H. Comprehensive analysis of SQOR involvement in ferroptosis resistance of pancreatic ductal adenocarcinoma in hypoxic environments. Front Immunol 2025; 16:1513589. [PMID: 40375994 PMCID: PMC12078260 DOI: 10.3389/fimmu.2025.1513589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 04/07/2025] [Indexed: 05/18/2025] Open
Abstract
Introduction Pancreatic ductal adenocarcinoma (PDAC) exhibits higher hypoxia level than most solid tumors, and the presence of intratumoral hypoxia is associated with a poor prognosis. However, the identification of hypoxia levels based on pathological images, and the mechanisms regulating ferroptosis resistance, remain to be elucidated. The objective of this study was to construct a deep learning model to evaluate the hypoxia characteristics of PDAC and to explore the role of Sulfide quinone oxidoreductase (SQOR) in hypoxia-mediated ferroptosis resistance. Methods Multi-omics data were integrated to analyze the correlation between hypoxia score of PDAC, SQOR expression and prognosis, and ferroptosis resistance level. A deep learning model of Whole Slide Images (WSIs) were constructed to predict the hypoxia level of patients. In vitro hypoxia cell models, SQOR knockdown experiments and nude mouse xenograft models were used to verify the regulatory function of SQOR on ferroptosis. Results PDAC exhibited significantly higher hypoxia levels than normal tissues, correlating with reduced overall survival in patients. In slide level, our deep learning model can effectively identify PDAC hypoxia levels with good performance. SQOR was upregulated in tumor tissues and positively associated with both hypoxia score and ferroptosis resistance. SQOR promotes the malignant progression of PDAC in hypoxic environment by enhancing the resistance of tumor cells to ferroptosis. SQOR knockdown resulted in decreased cell viability, decreased migration ability and increased MDA level under hypoxic Ersatin induced conditions. Furthermore, SQOR inhibitor in combination with ferroptosis inducer has the potential to inhibit tumor growth in vivo in a synergistic manner. Discussion This study has established a hypoxia detection model of PDAC based on WSIs, providing a new tool for clinical evaluation. The study revealed a new mechanism of SQOR mediating ferroptosis resistance under hypoxia and provided a basis for targeted therapy.
Collapse
Affiliation(s)
- Quan Lin
- Department of Hepatobiliary Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shiwei Guan
- Department of Hepatobiliary Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Minghui Peng
- Department of Hepatobiliary Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kailun Zhang
- Department of Hepatobiliary Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hewei Zhang
- Department of Hepatobiliary Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Taoming Mo
- Department of Pathology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Haibo Yu
- Department of Hepatobiliary Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
22
|
Yamane K, Anazawa T, Nagai K, Kasai Y, Masui T, Izuwa A, Kurahashi K, Ishida S, Ogiso S, Yoshimura M, Iwai T, Matsubara J, Fukuda A, Isoda H, Hidaka Y, Ibi Y, Hatano E. Neoadjuvant Chemoradiotherapy Using Moderately Hypofractionated Intensity-Modulated Radiotherapy Versus Upfront Surgery for Resectable Pancreatic Cancer: A Retrospective Cohort Study. Ann Surg Oncol 2025; 32:3603-3613. [PMID: 39893341 PMCID: PMC11976822 DOI: 10.1245/s10434-025-16956-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/16/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND The efficacy of neoadjuvant chemoradiotherapy for resectable pancreatic ductal adenocarcinoma (R-PDAC) remains unclear. This study was designed to evaluate neoadjuvant chemoradiotherapy by using intensity-modulated radiotherapy (NAC-IMRT) for R-PDAC compared with upfront surgery (UpS). METHODS Among 198 patients with R-PDAC who were indicated for resection between 2013 and 2021, 130 were included in this study after excluding patients who underwent neoadjuvant chemotherapy and did not meet the NAC-IMRT criteria (Eligible set). NAC-IMRT was planned for 58 patients, and UpS was planned for 72 patients. Additionally, in 105 patients who could undergo the planned treatment (As-treated set), the surgical, pathological, and oncological outcomes were evaluated. RESULTS In the Eligible set, median overall survival (OS) was 50.5 months with NAC-IMRT and 34.7 months with UpS and progression-free survival was 20.4 months with NAC-IMRT and 13.9 months with UpS. In the As-treated set, OS was longer in the NAC-IMRT group (66.7 months vs. 34.7 months, p = 0.007). On multivariate analysis, NAC-IMRT was identified as an independent factor for better OS (hazard ratio 0.617, 95% confidence interval 0.382-0.995, p = 0.047, in the Eligible set). The incidence of postoperative complications did not show a difference between the two groups, and NAC-IMRT suppressed local tumor invasion, including lymphatic, venous, perineural invasion, and lymph node metastases. CONCLUSIONS NAC-IMRT may offer superior survival outcomes and manageable toxicity in R-PDAC patients compared with upfront surgery. This study supports the efficacy and safety of NAC-IMRT and recommends its consideration in R-PDAC treatment protocols.
Collapse
Affiliation(s)
- Kei Yamane
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takayuki Anazawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Kazuyuki Nagai
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yosuke Kasai
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshihiko Masui
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Surgery, Kurashiki Central Hospital, Okayama, Japan
| | - Aya Izuwa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koki Kurahashi
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Ishida
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Ogiso
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Michio Yoshimura
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiro Iwai
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junichi Matsubara
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyoshi Isoda
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yu Hidaka
- Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yumiko Ibi
- Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Etsuro Hatano
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
23
|
Zhang R, Danshiitsoodol N, Noda M, Yonezawa S, Kanno K, Sugiyama M. Stevia Leaf Extract Fermented with Plant-Derived Lactobacillus plantarum SN13T Displays Anticancer Activity to Pancreatic Cancer PANC-1 Cell Line. Int J Mol Sci 2025; 26:4186. [PMID: 40362423 PMCID: PMC12071683 DOI: 10.3390/ijms26094186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Pancreatic cancer is a highly malignant tumor that remains a significant global health burden. In this study, we demonstrated the anticancer potential of stevia leaf extract fermented with plant-derived Lactobacillus (L.) plantarum SN13T strain. Evaluation of antioxidant capacity (including DPPH and ABTS radical scavenging activities and H2O2-induced oxidative damage repair in HEK-293 cells), as well as cytotoxicity against pancreatic cancer cells (PANC-1) and non-cancerous human embryonic kidney (HEK-293), revealed that the fermented extract exhibited significantly enhanced antioxidant activity and cytotoxicity against PANC-1 cells while showing minimal toxicity to HEK-293 cells compared to the unfermented extract. Further, validation through clonogenic, migration, and wound-healing assays demonstrated that the fermented extract effectively inhibited the proliferation and migration of PANC-1 cells. The active compound in the fermented extract has been identified as chlorogenic acid methyl ester (CAME), with a concentration of 374.4 μg/mL. Flow cytometry analysis indicated that CAME significantly arrested PANC-1 cells in the G0/G1 phase and induced apoptosis. Furthermore, CAME upregulated the expression of pro-apoptotic genes Bax, Bad, Caspase-3/9, Cytochrome c, and E-cadherin, while downregulating the anti-apoptotic gene Bcl-2. These findings suggest that CAME exerts potent cytotoxic effects on PANC-1 cells by inhibiting cell proliferation and migration, arresting the cell cycle, and regulating apoptosis-related gene expression. In conclusion, stevia leaf extract fermented with L. plantarum SN13T, which contains CAME, may serve as a promising candidate for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Rentao Zhang
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (R.Z.); (N.D.); (M.N.)
| | - Narandalai Danshiitsoodol
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (R.Z.); (N.D.); (M.N.)
| | - Masafumi Noda
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (R.Z.); (N.D.); (M.N.)
| | - Sayaka Yonezawa
- Department of General Internal Medicine, Hiroshima University Hospital, Hiroshima University, Hiroshima 734-8551, Japan; (S.Y.); (K.K.)
| | - Keishi Kanno
- Department of General Internal Medicine, Hiroshima University Hospital, Hiroshima University, Hiroshima 734-8551, Japan; (S.Y.); (K.K.)
| | - Masanori Sugiyama
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (R.Z.); (N.D.); (M.N.)
| |
Collapse
|
24
|
Zhai C, Ding X, Mao L, Ge Y, Huang A, Yang F, Ding Y. MEF2A, MEF2C, and MEF2D as potential biomarkers of pancreatic cancer? BMC Cancer 2025; 25:775. [PMID: 40281485 PMCID: PMC12023379 DOI: 10.1186/s12885-025-14107-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND The myocyte enhancer factor-2 (MEF2) family genes were involved in the carcinogenesis and prognosis of multiple human tumors. The impact of MEF2s on the occurrences, progression, and clinical outcome of pancreatic cancer (PAAD) remains unknown. METHODS This study used the CCLE, HPA, EMBL-EBI, and GEPIA2 databases to study MEF2s expression in PAAD patients. We also investigated the relationship between MEF2s expression and methylation through the DiseaseMeth database, and used MEXPRESS to verify the association. Then we utilized the Kaplan-Meier Plotter and GEPIA2 databases to evaluate the prognostic value of MEF2s in PAAD. The cBioPortal database was used to explore the alteration features of MEF2s in PAAD. We then investigated the association between MEF2s expression, immune cells infiltration, and immune infiltration markers using the TIMER database. Finally, Metascape, STRING, and Cytoscape tools were used for functional enrichment analysis. RESULTS MEF2A, MEF2C, and MEF2D were found to be highly expressed in PAAD patients' tissues compared to normal tissues, whereas MEF2B expression did not show significant differential expression. In addition, the protein expression of MEF2A, MEF2C, and MEF2D was higher in PAAD tissues. Negative correlations were observed between the expression level of MEF2A, MEF2C, and MEF2D and the methylation levels in multiple sites. High expression of MEF2A was related to poor overall survival (p = 0.0071) and relapse-free survival (RFS) (p = 0.0089) of PAAD. High expression of MEF2C was associated with worse RFS of PAAD (p = 0.043). MEF2A was a Truncating mutation, and it was shown that the "G27Wfs*8" mutation point was distributed in the SRF-TF domain. Both MEF2C and MEF2D were a Missense mutation. MEF2A, MEF2C, and MEF2D expression was positively corresponded with five immune cells infiltration (CD8 + T cells, B-cells, neutrophils, macrophages, and dendritic cells), especially for CD8 + T cells and macrophages. Among the 20 pathways, hsa05140 (Leishmania infection), hsa04022 (cGMP-PKG signaling pathway), hsa05145 (Toxoplasmosis), hsa04371 (Apelin signaling pathway), and hsa04064 (NF-kappa B signaling pathway), were closely connected with the occurrence and development of PAAD. CONCLUSIONS Our results indicated that the overexpression of MEF2A, MEF2C, and MEF2D in patients with PAAD. MEF2A could be used as a prognostic biomarker for PAAD, MEF2C might be a potential oncogene for PAAD, and MEF2D had potential biological significance.
Collapse
Affiliation(s)
- Chunxia Zhai
- Department of Public Health, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, Jiangsu, 226011, China.
| | - Xiaorong Ding
- Department of Public Health, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, Jiangsu, 226011, China
| | - Liping Mao
- Department of Oncology, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, Jiangsu, 226011, China
| | - Yang Ge
- Department of Public Health, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, Jiangsu, 226011, China
| | - Anqi Huang
- Department of Public Health, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, Jiangsu, 226011, China
| | - Fan Yang
- Department of Public Health, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, Jiangsu, 226011, China
| | - Yi Ding
- Department of Public Health, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, Jiangsu, 226011, China.
| |
Collapse
|
25
|
Noversa de Sousa R, Carqueja I, Sá Lima A, Guimarães F. Pancreatic cancer unveiled by multisystemic manifestations. BMJ Case Rep 2025; 18:e264969. [PMID: 40280580 DOI: 10.1136/bcr-2025-264969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025] Open
Abstract
Pancreatic cancer is characterised by its aggressive progression, late-stage diagnosis and poor prognosis. This case report illustrates the intricate interplay between pancreatic adenocarcinoma, disseminated intravascular coagulation with diffuse alveolar haemorrhage and bone metastasis in an elderly woman. Presenting with asthenia, significant hip pain and haemoptysis, her work-up revealed multiple supradiaphragmatic adenopathies and infradiaphragmatic adenopathies, pancreatic parenchymal heterogeneity and lytic bone lesions. Despite extensive diagnostic efforts, a diagnosis of adenocarcinoma of probable pancreatobiliary origin was established only at an advanced stage, leading to a fatal outcome within a week. The patient experienced rapid clinical deterioration, characterised by refractory respiratory failure, haematological dysfunction and circulatory shock, highlighting the aggressive course of pancreatic cancer and its complications. This case underscores the diagnostic challenges and the critical need for early recognition and tailored strategies in managing complex presentations of pancreatic cancer, especially with rare and severe systemic manifestations.
Collapse
Affiliation(s)
- Rita Noversa de Sousa
- Internal Medicine, Pedro Hispano Hospital, Matosinhos Local Health Unit, Matosinhos, Portugal
| | - Inês Carqueja
- Intensive Care Unit, Pedro Hispano Hospital, Matosinhos Local Health Unit, Matosinhos, Portugal
| | - Andreia Sá Lima
- Internal Medicine, Pedro Hispano Hospital, Matosinhos Local Health Unit, Matosinhos, Portugal
| | - Filipa Guimarães
- Intensive Care Unit, Pedro Hispano Hospital, Matosinhos Local Health Unit, Matosinhos, Portugal
| |
Collapse
|
26
|
Mu D, Chen B, Liu X, Zheng S, Zhang Y, Ni H, Zhou D. Exploring the potential mechanisms of Da ChaiHu decoction against pancreatic cancer based on network pharmacology prediction and molecular docking approach. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04107-w. [PMID: 40266298 DOI: 10.1007/s00210-025-04107-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 03/25/2025] [Indexed: 04/24/2025]
Abstract
Da ChaiHu decoction (DCHD) is used in Chinese medicine to treat pancreatic cancer (PC), but its exact mechanism is not known. The aim of this study was to investigate the main active ingredients and specific mechanisms of DCHD against PC. Firstly, the active ingredients and targets of DCHD and PC-related targets were searched from the TCMSP, DrugBank, NCBI and GeneCards databases, respectively. The intersected targets of both were then taken to construct a PPI network using STRING, and this network was visualized by Cytoscape 3.8.2. GO and KEGG enrichment analyses of the intersected targets were performed using R 4.2.1 "clusterProfiler", "enrichplot", and "ggplot2" packages. Molecular docking was performed utilizing MOE software to detect the binding capacity between compounds and targets. Cell proliferation, apoptosis, invasion and migration were examined through a CCK8 kit, Muse® Cell Analyzer, transwell and wound healing experiment, respectively. The expression levels of five core targets were assessed by RT-qPCR in PANC-1 cells treated with stigmasterol. Molecular dynamic simulations analysis was conducted to analyze the binding affinities and modes of interaction between molecules and stigmasterol using the GROMACS 5.1.4 program package. In this study, 141 common targets of DCHD and PC were obtained. GO-MF items indicated that DCHD exerts its effects on PC primarily by influencing the binding activity of DNA-binding transcription factors. The KEGG analysis revealed that these genes were implicated in various signaling pathways, including the IL-17 signaling pathway and the PI3K/Akt signaling pathway. Stigmasterol was chosen as the final ingredient for subsequent investigation due to its derivation from herb (Da ChaiHu), its encompassment of more common targets, and the scarcity of existing research on its role in PC. The results of molecular docking and Molecular dynamic simulations analysis showed that stigmasterol had good binding activity with BCL2, and ICAM1. In vitro experiments suggested that stigmasterol could effectively inhibit the proliferation, invasion and migration of PANC-1 cells, and promote cell apoptosis. Moreover, stigmasterol treatment led to the reduced expression of AKT1, HIF1A, BCL2, IL1B, and ICAM1. This study is the first to reveal the main active components and potential mechanisms of DCHD against PC, which provides a theoretical basis for studying the role of DCHD in the treatment of PC. Especially, the anti-PC mechanism of active compound stigmasterol might be associated with inhibiting proliferation, invasion and migration and accelerating apoptosis. Furthermore, five targets (AKT1, HIF1A, BCL2, IL1B, and ICAM1) were identified as key targets of stigmasterol, and the mRNA expressions of these genes were down-regulated by stigmasterol through in vitro experiments.
Collapse
Affiliation(s)
- Dong Mu
- Department of Gastroenterology and Hepatology, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Beijin Chen
- Department of Gastroenterology and Hepatology, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Xiaoli Liu
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, China
| | - Shumei Zheng
- Department of Gastroenterology and Hepatology, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Yong Zhang
- Department of Gastroenterology and Hepatology, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Hua Ni
- Department of Gastroenterology and Hepatology, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Dejiang Zhou
- Department of Gastroenterology and Hepatology, The General Hospital of Western Theater Command, Chengdu, 610083, China.
- , Chengdu, China.
| |
Collapse
|
27
|
Chen X, Sun S, Zhao J, Yu S, Chen J, Chen X. Tumor-stroma ratio combined with PD-L1 identifies pancreatic ductal adenocarcinoma patients at risk for lymph node metastases. Br J Cancer 2025:10.1038/s41416-025-03019-z. [PMID: 40246986 DOI: 10.1038/s41416-025-03019-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Pathological examination of lymph node metastasis (LNM) is crucial for treating pancreatic ductal adenocarcinoma (PDAC). Although the tumour stroma is correlated with prognosis in multiple solid tumors, its role in detecting LNM in PDAC is unclear. Thus, this study aimed to investigate the relationship of tumor-stroma ratio (TSR) with LNM, survival and mutational profile in PDAC. METHODS In this multicenter retrospective study, we examined molecular and clinicopathologic features of 737 PDAC patients from 5 independent cohorts, including surgically resected and endoscopic ultrasound fine-needle aspiration (EUS-FNA) biopsy specimens. TSR was evaluated on hematoxylin and eosin-stained slides and classified as stroma-low (<50% stroma) or stroma-high (≥50% stroma). RESULTS Compared to TSR-high cases, TSR-low cases were significantly associated with LNM (P < 0.001). TSR could accurately distinguish patients with and without LNM with an area under curve (AUC) of 0.749, with the sensitivity and specificity of 76.5% and 71.6%, respectively. This accuracy of TSR for identifying LNM was further increased by adding other factors including PD-L1 expression or pretreatment serum CA19-9 levels. TSR showed similar levels of accuracy in analysis of resected tumor specimens and EUS-FNA biopsies. Moreover, we found that TSR could also identify residual nodal involvement after neoadjuvant therapy (NAT) using pretreatment EUS-FNA biopsy samples. Heterogeneous genetic alterations were observed between TSR-low and TSR-high subgroups. TSR was identified as an independent predictor of LNM and worse disease-free survival. Major findings were all reproducible in validation, EUS-FNA biopsy, and pre-treatment NAT EUS-FNA biopsy cohorts. CONCLUSIONS TSR served as a robust and reproducible biomarker that identifies patients at risk for LNM. TSR might be used to select treatment and management strategies for PDAC patients.
Collapse
Affiliation(s)
- Xianlong Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shanyue Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jiapeng Zhao
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuangni Yu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Jie Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Xinyuan Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
28
|
Liu W, Rao L, Qiao Z, Wang G, Li B, Shen G. Global disparities in the burden of pancreatic cancer (1990-2021): insights from the 2021 Global Burden of Disease study. BMC Cancer 2025; 25:722. [PMID: 40247202 PMCID: PMC12007340 DOI: 10.1186/s12885-025-14110-2] [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: 11/16/2024] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is a highly lethal malignancy, ranking seventh among cancer-related deaths worldwide. This study utilizes data from the 2021 Global Burden of Disease (GBD) study to examine the global burden of PC and associated health inequalities from 1990 to 2021, with a focus on key risk factors such as obesity, high fasting plasma glucose, and the Socio-Demographic Index (SDI). METHODS Disability-Adjusted Life Years (DALYs) for PC were estimated using GBD 2021 data. The analysis incorporated SDI, age, gender, and major risk factors, including obesity and high fasting plasma glucose. Descriptive statistics and visualizations, such as age-sex pyramids and geographic maps, were employed to assess global, regional, and national burdens. Health disparities were quantified using the Concentration Index (CI) and the Slope Index of Inequality (SII), with CI assessing relative health distribution by income and SII measuring absolute socioeconomic inequality. RESULTS Globally, PC-related DALYs rose from 1.76 million in 1990 to 4.25 million in 2021 (141.48% increase), with the age-standardized DALY rate up 11.57% to 48.71 (95% UI 23.43 to 74.33). The burden was highest in high SDI regions, while low SDI areas still faced elevated rates; transitional and developing economies showed the highest age-standardized DALY rates. The SII increased from 189.63 (95% CI 177.65 to 245.17) in 1990 to 321.17 (95% CI 294.48 to 379.722) in 2021, indicating widening socioeconomic disparities. CONCLUSION PC remains a significant global health challenge with growing socioeconomic and geographic disparities. Urgent action is needed to address modifiable risk factors (e.g., obesity, diabetes) through enhanced healthcare infrastructure, early detection, and treatment access in low SDI countries, alongside improved data systems and international collaboration.
Collapse
Affiliation(s)
- Wei Liu
- Department of Minimally Invasive Common Surgery, Suzhou Ninth People's Hospital, Xuzhou Medical University Suzhou Bay Clinical College, Suzhou, Jiangsu Province, China
| | - Li Rao
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhengguo Qiao
- Department of Gastroenterology, Suzhou Ninth People's Hospital, Xuzhou Medical University, Suzhou Bay Clinical College, Suzhou, Jiangsu Province, China
| | - Gang Wang
- Department of Minimally Invasive Common Surgery, Suzhou Ninth People's Hospital, Xuzhou Medical University Suzhou Bay Clinical College, Suzhou, Jiangsu Province, China
| | - Bin Li
- Department of Minimally Invasive Common Surgery, Suzhou Ninth People's Hospital, Xuzhou Medical University Suzhou Bay Clinical College, Suzhou, Jiangsu Province, China
| | - Genhai Shen
- Department of Minimally Invasive Common Surgery, Suzhou Ninth People's Hospital, Xuzhou Medical University Suzhou Bay Clinical College, Suzhou, Jiangsu Province, China.
| |
Collapse
|
29
|
Liu J, Li H, Lin X, Xiong J, Wu G, Ding L, Lin B. Deciphering the heterogeneity of epithelial cells in pancreatic ductal adenocarcinoma: implications for metastasis and immune evasion. World J Surg Oncol 2025; 23:144. [PMID: 40240899 PMCID: PMC12004766 DOI: 10.1186/s12957-025-03793-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
OBJECTIVE This study examines the cellular heterogeneity of epithelial cells within pancreatic ductal adenocarcinoma (PDAC) and their contributions to tumor progression, metastasis, and immunosuppressive interactions using single-cell RNA sequencing. METHODS Single-cell RNA-sequencing data from two datasets (GSE154778 and GSE158356) were integrated using the Harmony algorithm, followed by quality control, clustering, and differential gene expression analysis. Distinct subpopulations of epithelial cells were identified, and their gene expression profiles were analyzed. To assess the malignancy of these subpopulations, single-cell copy number variation (CNV) analysis and trajectory analysis were conducted. Additionally, intercellular communication was examined using the CellChat platform. RESULTS The analysis revealed pronounced heterogeneity among PDAC epithelial cells, with specific subpopulations exhibiting distinct roles in tumor proliferation, extracellular matrix remodeling, and metastatic dissemination. Subpopulations 4 and 6 were characterized by increased CNV levels and a more malignant phenotype, suggesting an enhanced capacity for metastasis. Single-cell trajectory analysis, along with CellChat, mapped the temporal evolution of epithelial cells, identifying key regulatory genes such as DCBLD2 and JUN. A prognostic model incorporating five key genes, including KLF6, was developed and demonstrated strong predictive accuracy for patient outcomes. Notably, KLF6 emerged as a critical prognostic marker associated with immune modulation, particularly through interactions with M2 macrophages. CONCLUSION The study highlights the pronounced heterogeneity of epithelial cells in PDAC and their distinct contributions to tumor progression, metastasis, and immune modulation. Through single-cell transcriptomic and CNV analyses, we identified epithelial subpopulations with varying malignant potentials and distinct interactions with the tumor microenvironment. Among these, KLF6 emerged as a key regulator associated with immune modulation and metastasis. Our findings emphasize the significance of epithelial cell heterogeneity in shaping pancreatic cancer progression. These insights provide a foundation for future investigations into novel prognostic markers and therapeutic strategies.
Collapse
Affiliation(s)
- Jie Liu
- Department of Hepatopancreatobiliary Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Hui Li
- Department of Hepatopancreatobiliary Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Xiuyun Lin
- Department of Hepatopancreatobiliary Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Jiani Xiong
- Department of Hepatopancreatobiliary Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Guangfeng Wu
- Department of Hepatopancreatobiliary Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Lingyan Ding
- Department of Hepatopancreatobiliary Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Bin Lin
- Department of Orthodontics, Fujian Medical University Union Hospital, No. 29 of Xinquan Road, Gulou District, Fuzhou, 350001, China.
| |
Collapse
|
30
|
Wang X, Yang C, Liu Y, Wang J. SUMOylation substrate encoding genes as prognostic biomarkers in pancreatic ductal adenocarcinoma with functional assessment of SAF-B2. Front Pharmacol 2025; 16:1532658. [PMID: 40308776 PMCID: PMC12040899 DOI: 10.3389/fphar.2025.1532658] [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: 11/22/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is highly malignant with a poor prognosis, posing significant clinical challenges. SUMOylation, a reversible post-translational modification, plays a critical role in tumor progression, yet its prognostic significance in PDAC remains unclear. Methods We assessed SUMOylation expression patterns and function in PDAC using Western blot and the SUMOylation inhibitor TAK-981. Differentially expressed SUMOylation substrate encoding genes (DE-SSEGs) were identified from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression Project (GTEx) datasets. A SUMOylation-based prognostic model, Sscore, was constructed using LASSO and Cox regression. Additional analyses included somatic mutation, immune infiltration, TIDE, drug sensitivity, and single-cell RNA sequencing. The role of SAFB2 in PDAC was validated in vitro. Results PDAC cells showed elevated SUMOylation, and its inhibition reduced cell proliferation. The Sscore model, based on DE-SSEGs (CDK1, AHNAK2, SAFB2), predicted overall survival and correlated with genome variation, immune infiltration, and drug sensitivity. Single-cell analysis further confirmed a link between high Sscore and malignancy. SAFB2, identified as a pivotal gene within the Sscore model, was significantly downregulated in PDAC tissues and cell lines; its overexpression was shown to inhibit PDAC cell proliferation, migration, and invasion by suppressing the Wnt/β-Catenin signaling pathway. Conclusion This study underscores the role of SUMOylation in PDAC and introduces the Sscore as a prognostic tool. SAFB2 is identified as a potential tumor suppressor, offering new therapeutic targets for PDAC.
Collapse
Affiliation(s)
| | | | | | - Jian Wang
- Department of Hepatobiliary and Pancreatic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
31
|
Zare-Mehrjardi MJ, Hatami-Araghi M, Jafari-Khorchani M, Oushyani Roudsari Z, Taheri-Anganeh M, Abdolrahmat M, Ghasemi H, Aiiashi S. RNA biosensors for detection of pancreatic cancer. Clin Chim Acta 2025; 571:120237. [PMID: 40081786 DOI: 10.1016/j.cca.2025.120237] [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: 12/12/2024] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Pancreatic cancer is recognized as one of the most lethal types of cancer globally, characterized by a high mortality rate and a bleak prognosis, which greatly contributes to cancer-related deaths. Forecasts suggest that by 2030, pancreatic cancer will exceed other cancer types in prevalence. The disease presents considerable difficulties owing to the lack of prominent symptoms in its early stages, restricted options for early detection, rapid progression, and unfavorable outcomes. Presently, traditional methods for diagnosing pancreatic cancer primarily rely on imaging techniques. However, these methods often entail significant costs, require considerable time, and necessitate specialized skills for both operating the equipment and interpreting the resulting images. To overcome these obstacles, the use of biosensors has been proposed as a potentially valuable tool for the early detection of pancreatic cancer. MicroRNAs (miRs), a type of small non-coding RNA molecules, have emerged as highly sensitive molecular diagnostic tools that have the potential to function as precise indicators for a range of diseases, including cancer. Biosensors have been suggested as a potential solution for tackling these challenges, offering a promising approach for the early detection of pancreatic cancer. Small non-coding RNA molecules known as MicroRNAs (miRs) have become recognized as extremely sensitive molecular diagnostic tools and can act as precise biomarkers for different diseases, such as cancer. Moreover, this manuscript presents a thorough summary of the latest innovations in nano-biosensors that have been specifically developed for the identification of non-coding RNAs related to pancreatic cancer.
Collapse
Affiliation(s)
| | - Mahtab Hatami-Araghi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Majid Jafari-Khorchani
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Oushyani Roudsari
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Mona Abdolrahmat
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hassan Ghasemi
- Research Center for Environmental Contaminants (RCEC), Abadan University of Medical Sciences, Abadan, Iran.
| | - Saleh Aiiashi
- Abadan University of Medical Sciences, Abadan, Iran.
| |
Collapse
|
32
|
Dai X, Xi M, Li J. Cancer metastasis: molecular mechanisms and therapeutic interventions. MOLECULAR BIOMEDICINE 2025; 6:20. [PMID: 40192949 PMCID: PMC11977077 DOI: 10.1186/s43556-025-00261-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 04/10/2025] Open
Abstract
The metastatic cascade is a complicated process where cancer cells travel across multiple organs distant from their primary site of onset. Despite the wide acceptance of the 'seed and soil' theory, mechanisms driving metastasis organotropism remain mystery. Using breast cancer of different subtypes as the disease model, we characterized the 'metastatic profile of cancer cells' and the 'redox status of the organ microenvironment' as the primary determinants of cancer metastasis organotropism. Mechanically, we identified a positive correlation between cancer metabolic plasticity and stemness, and proposed oxidative stress as the selection power of cancer cells succeeding the metastasis cascade. Therapeutically, we proposed the use of pro-oxidative therapeutics in ablating cancer cells taking advantages of this fragile moment during metastasis. We comprehensively reviewed current pro-oxidative strategies for treating cancers that cover the first line chemo- and radio-therapies, approaches relying on naturally existing power including magnetic field, electric field, light and sound, nanoparticle-based anti-cancer composites obtained through artificial design, as well as cold atmospheric plasma as an innovative pro-oxidative multi-modal modality. We discussed possible combinations of pro-oxidative approaches with existing therapeutics in oncology prior to the forecast of future research directions. This paper identified the fundamental mechanics driving metastasis organotropism and proposed intervention strategies accordingly. Insights provided here may offer clues for the design of innovative solutions that may open a new paradigm for cancer treatment.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China.
| | - Ming Xi
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Jitian Li
- Molecular Biology Lab, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Henan Province, Zhengzhou, 450000, China
| |
Collapse
|
33
|
Liang X, Tian R, Li T, Wang H, Qin Y, Qian M, Fan J, Wang D, Cui HY, Jiang J. Integrative insights into the role of CAV1 in ketogenic diet and ferroptosis in pancreatic cancer. Cell Death Discov 2025; 11:139. [PMID: 40180904 PMCID: PMC11968908 DOI: 10.1038/s41420-025-02421-z] [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: 08/04/2024] [Revised: 02/26/2025] [Accepted: 03/20/2025] [Indexed: 04/05/2025] Open
Abstract
Pancreatic cancer exhibits high mortality rates with limited therapeutic options. Emerging evidence suggests that the ketogenic diet may act as adjuvant therapy by triggering ferroptosis in cancer cells, though the underlying molecular mechanisms remain unclear. This study aims to investigate the molecular mechanisms linking ketogenic metabolism and ferroptosis, with an emphasis on key regulatory proteins. We demonstrated that pancreatic adenocarcinoma (PAAD) tissues significantly enhanced ketogenic and ferroptosis phenotypes compared to normal tissues, both correlating with poorer patient prognosis. These phenotypes showed strong interdependence mediated by CAV1. In the pancreatic tumor microenvironment, CAV1 was predominantly expressed in tumor cells. Through in vitro cell experiments, we clarified that Na-OHB downregulated CAV1 expression in pancreatic cancer cells, inhibiting the transcription of the CAV1/AMPK/NRF2 downstream ferroptosis-protective genes SLC7A11 and SLC40A1. Additionally, we demonstrated the interaction between CAV1 and SLC7A11 molecules; when CAV1 was downregulated, it affected the stability of SLC7A11, leading to the ubiquitination and degradation of the translated SLC7A11 protein. Through these dual mechanisms, Na-OHB caused Fe2+ overload, lipid peroxidation accumulation, and oxidative stress in pancreatic cancer cells, ultimately triggering ferroptosis. In ketogenic diet-fed tumor-bearing mouse models, we also observed a significant increase in lipid peroxidation and other related biomarkers, while CAV1 and SLC7A11 levels were markedly decreased compared to the normal diet group. Our findings identify CAV1 as a pivotal molecular link between ketogenic metabolism and ferroptosis in pancreatic cancer. The multi-level regulatory axis involving CAV1-mediated transcriptional regulation and post-translational modifications provides mechanistic insights into ketogenic diet-induced ferroptosis, suggesting potential therapeutic targets for pancreatic cancer adjuvant treatment.
Collapse
Affiliation(s)
- Xue Liang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi, 710032, China
| | - Ruofei Tian
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi, 710032, China
| | - Ting Li
- Cardiovascular Surgery Department, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, China
| | - Hao Wang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi, 710032, China
| | - Yifei Qin
- Institutes of Biomedicine and Department of Cell Biology, Jinan University, Guangzhou, 510632, China
| | - Meirui Qian
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi, 710032, China
| | - Jing Fan
- Air Force Hospital of the Northern Theater Command of the People's Liberation Army of China, Shenyang, Liaoning, 110003, China
| | - Dan Wang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi, 710032, China
| | - Hong-Yong Cui
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi, 710032, China
| | - Jianli Jiang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, Shaanxi, 710032, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
34
|
Michalet M, Valenzuela G, Nougaret S, Tardieu M, Azria D, Riou O. Development of Multiparametric Prognostic Models for Stereotactic Magnetic Resonance Guided Radiation Therapy of Pancreatic Cancers. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00282-2. [PMID: 40185208 DOI: 10.1016/j.ijrobp.2025.03.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/07/2025]
Abstract
PURPOSE Stereotactic magnetic resonance guided adaptive radiation therapy (SMART) is a new option for local treatment of unresectable pancreatic ductal adenocarcinoma, showing interesting survival and local control (LC) results. Despite this, some patients will experience early local and/or metastatic recurrence leading to death. We aimed to develop multiparametric prognostic models for these patients. METHODS AND MATERIALS All patients treated in our institution with SMART for an unresectable pancreatic ductal adenocarcinoma between October 21, 2019, and August 5, 2022 were included. Several initial clinical characteristics as well as dosimetric data of SMART were recorded. Radiomics data from 0.35-T simulation magnetic resonance imaging were extracted. All these data were combined to build prognostic models of overall survival (OS) and LC using machine learning algorithms. RESULTS Eighty-three patients with a median age of 64.9 years were included. A majority of patients had a locally advanced pancreatic cancer (77%). The median OS was 21 months after SMART completion and 27 months after chemotherapy initiation. The 6- and 12-month post-SMART OS was 87.8% (IC95%, 78.2%-93.2%) and 70.9% (IC95%, 58.8%-80.0%), respectively. The best model for OS was the Cox proportional hazard survival analysis using clinical data, with a concordance index inverse probability of censoring weighted of 0.87. Tested on its 12-month OS prediction capacity, this model had good performance (sensitivity 67%, specificity 71%, and area under the curve 0.90). The median LC was not reached. The 6- and 12-month post-SMART LC was 92.4% [IC95%, 83.7%-96.6%] and 76.3% [IC95%, 62.6%-85.5%], respectively. The best model for LC was the component-wise gradient boosting survival analysis using clinical and radiomics data, with a concordance index inverse probability of censoring weighted of 0.80. Tested on its 9-month LC prediction capacity, this model had good performance (sensitivity 50%, specificity 97%, and area under the curve 0.78). CONCLUSIONS Combining clinical and radiomics data in multiparametric prognostic models using machine learning algorithms showed good performance for the prediction of OS and LC. External validation of these models will be needed.
Collapse
Affiliation(s)
- Morgan Michalet
- University Federation of Radiation Oncology of Mediterranean Occitanie, Montpellier Cancer Institute, Montpellier University, Montpellier, France; Montpellier Cancer Research Institute, Montpellier University, Montpellier, France.
| | - Gladis Valenzuela
- Montpellier Cancer Research Institute, Montpellier University, Montpellier, France
| | - Stéphanie Nougaret
- Montpellier Cancer Research Institute, Montpellier University, Montpellier, France
| | - Marion Tardieu
- Montpellier Cancer Research Institute, Montpellier University, Montpellier, France
| | - David Azria
- University Federation of Radiation Oncology of Mediterranean Occitanie, Montpellier Cancer Institute, Montpellier University, Montpellier, France; University Federation of Radiation Oncology of Mediterranean Occitanie, Institut de Cancérologie du Gard, Centre Hospitalier Universitaire Carémeau, Nîmes, France; Montpellier Cancer Research Institute, Montpellier University, Montpellier, France
| | - Olivier Riou
- University Federation of Radiation Oncology of Mediterranean Occitanie, Montpellier Cancer Institute, Montpellier University, Montpellier, France; Montpellier Cancer Research Institute, Montpellier University, Montpellier, France
| |
Collapse
|
35
|
Li YC, Zhang L, Wang YT, Hu H, Zhang ZY, Nie QQ, Zuo CJ. Role of EFNAs in Shaping the Tumor Immune Microenvironment and Their Impact on Pancreatic Adenocarcinoma Prognosis. Cancer Manag Res 2025; 17:693-712. [PMID: 40190415 PMCID: PMC11972607 DOI: 10.2147/cmar.s502401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
Purpose Due to the highly heterogeneous and immunosuppressed tumor microenvironment (TME), pancreatic adenocarcinoma (PAAD) has limited therapeutic options and an abysmal prognosis. Ephrin A 1-5 (EFNA1-5) have been shown to regulate tumorigenesis and metastasis in various cancers, but its role in PAAD remains unclear. Methods We comprehensively analyzed EFNA gene expression levels in pan-cancer and PAAD using the GEPIA and HPA databases. Then, we assessed the prognostic value of EFNA1-5 using the Kaplan-Meier plotter and nomogram model. Further exploration of the association of EFNA1-5 with clinicopathological features of PAAD used information from the UALCAN database, and the TIMER dataset was used to reveal the correlation between EFNA1-5 and the tumor immune microenvironment (TIME) of pancreatic cancer. In addition, cBioPortal Databases, GSEA, and GSCALite were used to explore gene changes, protein interactions, and biological functions. Finally, the oncogenic effect of EFNA5 was verified in vivo and in vitro. Results The expression levels of EFNA1-5 were significantly upregulated in PAAD. The expression of EFNA1/3/4/5 were significantly associated with overall survival (OS) and relapse-free survival (RFS) in PAAD patients. The high expression of EFNA2-5 were related to poor clinical features, such as higher tumor stage or grade and a wider range of lymph node metastasis. EFNA1-5 were closely associated with immune cell infiltration, CAFs, and MDSCs expression. Furthermore, EFNA5 is an independent risk factor for poor prognosis in PAAD patients, and it can promote the malignant progression of pancreatic cancer in vitro and in vivo. Conclusion Differential expression of EFNA1-5 is associated with TIME in pancreatic cancer, predicts different survival outcomes, and maybe a novel prognostic marker reflecting an immunosuppressive state and a potential therapeutic target.
Collapse
Affiliation(s)
- Yu-Chao Li
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Lu Zhang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Yi-Ting Wang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Hao Hu
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Ze-Yu Zhang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Qian-Qian Nie
- Department of Central Laboratory, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Chang-Jing Zuo
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
36
|
Ding Y, Yu Y. Therapeutic potential of flavonoids in gastrointestinal cancer: Focus on signaling pathways and improvement strategies (Review). Mol Med Rep 2025; 31:109. [PMID: 40017144 PMCID: PMC11884236 DOI: 10.3892/mmr.2025.13474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
Flavonoids are a group of polyphenolic compounds distributed in vegetables, fruits and other plants, which have considerable antioxidant, anti‑tumor and anti‑inflammatory activities. Several types of gastrointestinal (GI) cancer are the most common malignant tumors in the world. A large number of studies have shown that flavonoids have inhibitory effects on cancer, and they are recognized as a class of potential anti‑tumor drugs. Therefore, the present review investigated the molecular mechanisms of flavonoids in the treatment of different types of GI cancer and summarized the drug delivery systems commonly used to improve their bioavailability. First, the classification of flavonoids and the therapeutic effects of various flavonoids on human diseases were briefly introduced. Then, to clarify the mechanism of action of flavonoids on different types of GI cancer in the human body, the metabolic process of flavonoids in the human body and the associated signaling pathways causing five common types of GI cancer were discussed, as well as the corresponding therapeutic targets of flavonoids. Finally, in clinical settings, flavonoids have poor water solubility, low permeability and inferior stability, which lead to low absorption efficiency in vivo. Therefore, the three most widely used drug delivery systems were summarized. Suggestions for improving the bioavailability of flavonoids and the focus of the next stage of research were also put forward.
Collapse
Affiliation(s)
- Ye Ding
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yong Yu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
37
|
Sanghvi G, Roopashree R, Kashyap A, Sabarivani A, Ray S, Bhakuni PN. KIFC1 in cancer: Understanding its expression, regulation, and therapeutic potential. Exp Cell Res 2025; 447:114510. [PMID: 40058447 DOI: 10.1016/j.yexcr.2025.114510] [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: 01/10/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025]
Abstract
Kinesins are a family of motor proteins essential for intracellular transport and cellular dynamics, with kinesin family member C1 (KIFC1) emerging as a key regulator of cancer progression. Recent studies highlight KIFC1's crucial role in mitotic spindle assembly, chromosome segregation, and cell migration-processes frequently dysregulated in cancer. Its involvement in promoting malignant cell proliferation and metastasis underscores its significance in tumor biology. In various cancer types, aberrant KIFC1 expression correlates with poor prognosis and aggressive phenotypes, suggesting its potential as a biomarker for disease severity. Mechanistically, KIFC1 influences signaling pathways linked to cell cycle regulation and programmed cell death, reinforcing its role in oncogenesis. Given its pivotal function in cancer cell dynamics, KIFC1 represents a promising therapeutic target. Strategies aimed at modulating its activity, including small molecules or RNA interference, could disrupt cancer cell viability and proliferation. The current review article highlights KIFC1's importance in cancer biology, advocating for further investigation into its mechanisms and the development of KIFC1-targeted therapies to enhance treatment efficacy and improve patient outcomes across various malignancies.
Collapse
Affiliation(s)
- Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, 360003, Gujarat, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Aditya Kashyap
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - A Sabarivani
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Pushpa Negi Bhakuni
- Department of Allied Science, Graphic Era Hill University, Bhimtal, Uttarakhand, 248002, India; Graphic Era Deemed to be University, Dehradun, Uttarakhand, India.
| |
Collapse
|
38
|
Qin C, Xu C, Zhu Z, Song X, Wang X, Xu W, Zhu M. A study of the association between Helicobacter pylori infection type and pancreatic cancer risk: A systematic review and meta‑analysis. Oncol Lett 2025; 29:174. [PMID: 39975953 PMCID: PMC11837465 DOI: 10.3892/ol.2025.14920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025] Open
Abstract
Pancreatic cancer is a highly invasive malignant tumor with a complex pathogenesis that makes early diagnosis challenging. The potential association between Helicobacter pylori infection and pancreatic cancer risk has been noted; however, the available results are still highly divergent. The aim of the present study was to systematically evaluate the association between different types of H. pylori infection and pancreatic cancer risk as well as to explore the possible causes. A systematic search was conducted using the PubMed, Embase and Cochrane Library databases up to August 2023. The literature quality was evaluated using the Newcastle-Ottawa Scale. All studies that met the criteria were included in the overall meta-analysis to calculate the odds ratios (ORs) and corresponding 95% confidence intervals (CIs). In addition, subgroup analyses were performed based on factors such as diagnostic criteria for H. pylori infection, study region, type of study design and CagA status. The effect of publication bias on the quantitative synthesis results was assessed using the trim-and-fill analysis, and sensitivity analyses were used to verify the robustness of the quantitative synthesis results. A total of 17 studies involving 67,910 participants, including 64,372 controls and 3,538 patients with pancreatic cancer, were included in the present study. The overall analysis showed that no significant association was observed between H. pylori infection and pancreatic cancer risk (OR, 1.15; 95% CI, 0.93-1.41). Further subgroup analyses, which did not consider the effects of study quality, diagnostic criteria, geographical distribution and the type of study design, did not produce new findings that contradicted the results of the overall analysis. CagA+ H. pylori infection did not significantly affect the risk of pancreatic cancer (OR, 0.95; 95% CI, 0.78-1.16), whereas CagA- H. pylori infection may be a possible risk factor for pancreatic cancer (OR, 1.24; 95% CI, 1.004-1.541). The H. pylori infection did not significantly increase the risk of pancreatic cancer. However, it is noteworthy that CagA- H. pylori infection could be a potential factor that elevated the risk of pancreatic cancer.
Collapse
Affiliation(s)
- Chao Qin
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Chonghe Xu
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, P.R. China
| | - Zhongqi Zhu
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Xixi Song
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Xin Wang
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Wei Xu
- Department of Blood Transfusion, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Mei Zhu
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| |
Collapse
|
39
|
Gao Y, Zandieh K, Zhao K, Khizanishvili N, Fazio PD, Yu X, Schulte L, Aillaud M, Chung HR, Ball Z, Meixner M, Bauer UM, Bartsch DK, Buchholz M, Lauth M, Nimsky C, Cook L, Bartsch JW. The long non-coding RNA NEAT1 contributes to aberrant STAT3 signaling in pancreatic cancer and is regulated by a metalloprotease-disintegrin ADAM8/miR-181a-5p axis. Cell Oncol (Dordr) 2025; 48:391-409. [PMID: 39412616 PMCID: PMC11996950 DOI: 10.1007/s13402-024-01001-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2024] [Indexed: 12/05/2024] Open
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers and several studies demonstrate that STAT3 has critical roles throughout the course of PDAC pathogenesis. METHODS TCGA, microarray, and immunohistochemistry data from a PDAC cohort were used for clinical analyses. Panc89 cells with ADAM8 knockout, re-expression of ADAM8 mutants, and Panc1 cells overexpressing ADAM8 were generated. Gene expression analyses of ADAM8, STAT3, long non-coding (lnc) RNA NEAT1, miR-181a-5p and ICAM1 were performed by quantitative PCR. Subcellular fractionation quantified NEAT1 expression in cytoplasm and nucleus of PDAC cell lines. Cell proliferation, scratch, and invasion assays were performed to detect growth rate, migration and invasion capabilities of cells. Gain and loss of function experiments were carried out to investigate the biological effects of lncRNA NEAT1 and miR-181a-5p on PDAC cells and downstream genes. Dual-luciferase reporter gene assay determined interaction and binding sites of miR-181a-5p in lncRNA NEAT1. Pull down assays, RNA binding protein immunoprecipitation (RIP), and ubiquitination assays explored the molecular interaction between lncRNA NEAT1 and STAT3. RESULTS High ADAM8 expression causes aberrant STAT3 signaling in PDAC cells and is positively correlated with NEAT1 expression. NEAT1 binding to STAT3 was confirmed and prevents STAT3 degradation in the proteasome as increased degradation of STAT3 was observed in ADAM8 knockout cells and cells treated with bortezomib. Furthermore, miRNA-181a-5p regulates NEAT1 expression by direct binding to the NEAT1 promoter. CONCLUSION ADAM8 regulates intracellular STAT3 levels via miR-181a-5p and NEAT1 in pancreatic cancer.
Collapse
Affiliation(s)
- Yutong Gao
- Department of Neurosurgery, Philipps-University Marburg, Baldingerstrasse, 35043, Marburg, Germany
| | - Kimia Zandieh
- Department of Neurosurgery, Philipps-University Marburg, Baldingerstrasse, 35043, Marburg, Germany
| | - Kai Zhao
- Department of Neurosurgery, Philipps-University Marburg, Baldingerstrasse, 35043, Marburg, Germany
| | - Natalia Khizanishvili
- Department of Visceral, Thoracic and Vascular Surgery, Philipps-University Marburg, Baldingerstrasse, 35033, Marburg, Germany
| | - Pietro Di Fazio
- Department of Visceral, Thoracic and Vascular Surgery, Philipps-University Marburg, Baldingerstrasse, 35033, Marburg, Germany
| | - Xiangdi Yu
- Department of Anesthesiology, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou University, Guiyang, Guizhou, 550000, China
| | - Leon Schulte
- Institute for Lung Research, Philipps-University Marburg, Hans-Meerwein-Strasse 2, 35043, Marburg, Germany
| | - Michelle Aillaud
- Institute for Lung Research, Philipps-University Marburg, Hans-Meerwein-Strasse 2, 35043, Marburg, Germany
| | - Ho-Ryun Chung
- Institute for Medical Bioinformatics and Biostatistics, Philipps-University Marburg, 35033, Marburg, Germany
| | - Zachary Ball
- Department of Chemistry, Rice University, Houston, TX, USA
| | - Marion Meixner
- Institute for Molecular Biology and Tumor Research (IMT), Philipps-University Marburg, Marburg, Germany
| | - Uta-Maria Bauer
- Institute for Molecular Biology and Tumor Research (IMT), Philipps-University Marburg, Marburg, Germany
| | - Detlef Klaus Bartsch
- Department of Visceral, Thoracic and Vascular Surgery, Philipps-University Marburg, Baldingerstrasse, 35033, Marburg, Germany
| | - Malte Buchholz
- Department of Gastroenterology, Endocrinology, Metabolism and Infection, Center for Tumor and Immunology (ZTI), Philipps-University Marburg, Marburg, Germany
| | - Matthias Lauth
- Institute for Molecular Biology and Tumor Research (IMT), Philipps-University Marburg, Marburg, Germany
| | - Christopher Nimsky
- Department of Neurosurgery, Philipps-University Marburg, Baldingerstrasse, 35043, Marburg, Germany
| | - Lena Cook
- Department of Neurosurgery, Philipps-University Marburg, Baldingerstrasse, 35043, Marburg, Germany
| | - Jörg W Bartsch
- Department of Neurosurgery, Philipps-University Marburg, Baldingerstrasse, 35043, Marburg, Germany.
| |
Collapse
|
40
|
Su D, Ruan Y, Shi Y, Cao D, Wu T, Dang T, Wang H, Xin Y, Ma M, Meng H, Liu C, Zhang Y. Molecular Subtyping and Genomic Profiling Expand Precision Medicine in KRAS Wild-Type Pancreatic Cancer. Cancer Sci 2025; 116:1094-1106. [PMID: 39833990 PMCID: PMC11967249 DOI: 10.1111/cas.16456] [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: 12/04/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease with poor prognosis and limited treatment options. While the majority of PDAC cases harbor KRAS mutations, approximately 8%-10% are KRAS wild-type (KRAS-WT). These KRAS-WT tumors often contain actionable mutations and gene fusions, making them more suitable for precision therapies. Identifying these molecular alterations is crucial for improving outcomes in this subset of patients. This retrospective study involved 34 patients with KRAS-WT PDAC. Genomic profiling was performed using next-generation sequencing (NGS) and RNA sequencing to detect mutations and fusions. Comparative analysis was conducted with TCGA-PAAD data, and immune infiltration was assessed using bioinformatic deconvolution methods. Targetable alterations were identified in multiple pathways. Key mutations included ATM (18%), PIK3CA (15%), and ROS1 (15%), while actionable gene fusions such as CCDC6-RET and ETV6-NTRK3 were present in 10.3% of patients. The gene mutations associated with homologous recombination deficiency (HRD) are predicted to increase sensitivity to platinum-based chemotherapy (p = 0.047). Tumors with epigenetic regulatory genes mutations (e.g., ARID1A, KMT2C/D) exhibited enhanced immune cell infiltration, highlighting potential responsiveness to immune checkpoint inhibitors (ICIs). Kinase fusions (NTRK and RET) were linked to response to larotinib and RET-specific inhibitors, respectively. KRAS-WT PDAC contains actionable mutations and fusions, offering significant potential for targeted and immune-based therapies. Further clinical studies are needed to validate these therapeutic approaches.
Collapse
Affiliation(s)
- Dan Su
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
| | - Yuli Ruan
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
- Clinical Research Center for Colorectal Cancer in HeilongjiangHarbinChina
| | - Yingfei Shi
- Department of Translational MedicineGenetron Health (Beijing) Technology, Co. Ltd.BeijingChina
| | - Dandan Cao
- Department of Translational MedicineGenetron Health (Beijing) Technology, Co. Ltd.BeijingChina
| | - Tong Wu
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
- Clinical Research Center for Colorectal Cancer in HeilongjiangHarbinChina
| | - Tianjiao Dang
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
| | - Hong Wang
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
| | - Yaqun Xin
- Department of Translational MedicineGenetron Health (Beijing) Technology, Co. Ltd.BeijingChina
| | - Ming Ma
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
| | - Hongxue Meng
- Department of PathologyHarbin Medical University Cancer HospitalHarbinChina
| | - Chao Liu
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
- Clinical Research Center for Colorectal Cancer in HeilongjiangHarbinChina
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical OncologyHarbin Medical University Cancer HospitalHarbinChina
- Key Laboratory of Tumor Immunology in HeilongjiangHarbinChina
- Clinical Research Center for Colorectal Cancer in HeilongjiangHarbinChina
| |
Collapse
|
41
|
Morand S, Rager L, Craig D, Nemunaitis A, Choucair K, Rao D, Stanbery L, Phinney RC, Walter A, Ghisoli M, Nemunaitis J. Clinical characterization and therapeutic targeting of fusion genes in oncology. Future Oncol 2025; 21:1249-1260. [PMID: 40128124 PMCID: PMC11988278 DOI: 10.1080/14796694.2025.2477974] [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: 10/15/2024] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
Gene fusions represent important oncogenic driver mutations resulting in aberrant cellular signaling. In up to 17% of all solid tumors at least one gene fusion can be identified. Precision therapy targeting fusion gene signaling has demonstrated effective clinical benefit. Advancements in clinically relevant next-generation sequencing and bioinformatic techniques have enabled expansion of therapeutic opportunity to subpopulations of patients with fusion gene expression. Clinically, tyrosine inhibitors have shown efficacy in treating fusion gene expressing cancers. Fusion genes are also clonal mutations, meaning it is a personal cancer target involving all cancer cells of that patient, not just a subpopulation of cancer cells within the cancer mass. Thus, both fusion signal disruption and immune signal targeting are effective therapeutic directions. This review discusses fusion gene targeting, therapeutic resistance, and molecular biomarkers.
Collapse
Affiliation(s)
- Susan Morand
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Lauren Rager
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Daniel Craig
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | | | - Khalil Choucair
- Department of Hematology/Oncology, Barbara Karmanos Cancer Institute,Wayne State University, Detroit, MI, USA
| | - Donald Rao
- Medical Affairs, Gradalis Inc, Dallas, TX, USA
| | - Laura Stanbery
- Taylor Cancer Research Center, Maumee, OH, USA
- Medical Affairs, Gradalis Inc, Dallas, TX, USA
| | - Richard C. Phinney
- Taylor Cancer Research Center, Maumee, OH, USA
- Department of Hematology/Oncology, Toledo Clinic Cancer Center, Maumee, OH, USA
| | - Adam Walter
- Medical Affairs, Gradalis Inc, Dallas, TX, USA
- Department of Gynecologic Oncology, Promedica Health System, Toledo, OH, USA
| | - Maurizio Ghisoli
- Department of Pediatric Hematology/Oncology, Texas Oncology, P.A, Dallas, TX, USA
| | - John Nemunaitis
- Taylor Cancer Research Center, Maumee, OH, USA
- Medical Affairs, Gradalis Inc, Dallas, TX, USA
- Department of Hematology/Oncology, Toledo Clinic Cancer Center, Maumee, OH, USA
| |
Collapse
|
42
|
Petrauskas V, Damaseviciute R, Gulla A. Pancreatic 3D Organoids and Microfluidic Systems-Applicability and Utilization in Surgery: A Literature Review. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:623. [PMID: 40282914 PMCID: PMC12028617 DOI: 10.3390/medicina61040623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/05/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025]
Abstract
Background: Pancreatic organoids are a rapidly advancing field of research with new discoveries being made every day. A literature review was performed to answer the question of how relevant 3D pancreatic organoids are for surgery. Materials and Methods: We started our investigation by identifying articles in PubMed within the last 5 years using the keywords (("pancreatic organoid", OR "organ-on-a-chip", OR "pancreatic chip" OR "3D culture methods") AND pancreatic surgery). Only English articles were included in this literature review. This literature review was performed in a non-systematic way; articles were chosen without a predetermined protocol of inclusion and were based on the aim of the review. Results and Conclusions: There are many promising innovations in the field of 3D cultures. Drug sensitivity testing in particular holds great potential for surgical application. For locally advanced PDAC, EUS-FNB obtained cancer tissue can be cultured as organoids, and after 4 weeks, neoadjuvant treatment could be adjusted for each patient individually. Utilizing this approach could increase the number of R0 resections and possibly cure the disease. Furthermore, microfluidic devices, as a platform for pancreatic islet pre-transplant evaluation or cultivation of beta cells derived from HiPSC in vitro, promise broad application of islet transplantation to T1DM patients in the near future.
Collapse
Affiliation(s)
- Vidas Petrauskas
- Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, LT-01131 Vilnius, Lithuania
| | - Ryte Damaseviciute
- Center of Visceral Medicine and Translational Research, Faculty of Medicine, Vilnius University, LT-01131 Vilnius, Lithuania
| | - Aiste Gulla
- Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, LT-01131 Vilnius, Lithuania
- Center of Visceral Medicine and Translational Research, Faculty of Medicine, Vilnius University, LT-01131 Vilnius, Lithuania
- Department of Surgery, George Washington University, Washington, DC 20052, USA
| |
Collapse
|
43
|
Cui R, Wang G, Hu R, Wang Y, Mu H, Song Y, Chen B, Jiang X. Prognostic and immunotherapeutic potential of disulfidptosis-associated signature in pancreatic cancer. Front Immunol 2025; 16:1568976. [PMID: 40207217 PMCID: PMC11979277 DOI: 10.3389/fimmu.2025.1568976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Disulfidptosis is a newly discovered formation of programmed cell death. However, the significance of disulfidptosis in pancreatic adenocarcinoma remains unclear. Our investigation aims to elucidate the significance of disulfidptosis in pancreatic ductal adenocarcinoma by integrating diverse datasets, including bulk RNA sequencing data, microarray profiles, single-cell transcriptome profiles, spatial transcriptome data, and biospecimens. Utilizing various bioinformatics tools, we screened disulfidptosis-related genes based on single-cell RNA sequencing profiles, subsequently validating them through enrichment analysis. An 8-gene disulfidptosis-related prognostic signature was established by constructing massive LASSO-Cox regression models and validated by multiple external PDAC cohorts. Evaluation methods, such as Kaplan-Meier curves, ROC curves, time-dependent ROC curves, and decision curve analysis, were employed to assess the prognostic signature's reliability. High disulfidptosis-related scores were associated with a poorer prognosis and diminished sensitivity to immune checkpoint blockade. Further investigation uncovered that the potential components of elevated DPS involve malignant tumor hallmarks, extensive interactions between myCAFs and tumor cells, and the exclusion of immune cells. Cell-cell communication analysis highlighted myCAFs' role in signaling, potentially influencing tumor cells towards increased malignancy through collagen, laminin, and FN1 signaling networks. Spatial transcriptome analysis confirmed the crosstalk between myCAFs and tumor cells. Biospecimens including 20 pairs of PDAC samples and adjacent normal tissues further demonstrated the robustness of DPS and its correlation with CAF markers. In conclusion, our study introduces a novel disulfidptosis-related signature with high efficacy in patient risk stratification, which has the ability to predict the sensitivity to immune checkpoint blockade.
Collapse
Affiliation(s)
- Ran Cui
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Gaoming Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Renhao Hu
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yongkun Wang
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huiling Mu
- Department of Biobank, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanxiang Song
- Department of Biobank, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bo Chen
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaohua Jiang
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
44
|
Shionoya K, Sofuni A, Mukai S, Tsuchiya T, Tanaka R, Tonozuka R, Yamamoto K, Nagai K, Matsunami Y, Kojima H, Minami H, Hirakawa N, Asano K, Yamaguchi Y, Hama K, Itoi T. Evaluating the Usefulness of the Blood Apolipoprotein A2 Isoform Index for Pancreatic Cancer Diagnosis. Cancers (Basel) 2025; 17:1071. [PMID: 40227633 PMCID: PMC11987948 DOI: 10.3390/cancers17071071] [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: 02/23/2025] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Early detection of pancreatic cancer using existing tumor markers is challenging, and novel biomarkers are needed. Apolipoprotein A2 (APOA2), which is not directly produced by tumors, may help detect pancreatic cancer through mechanisms distinct from carbohydrate antigen 19-9 (CA 19-9). This study aimed to evaluate the diagnostic performance of the APOA2-isoform (APOA2-i) Index in patients with pancreatic cancer. Methods: Serum levels of the APOA2-i Index and CA 19-9 were measured in 76 patients with pancreatic cancer (Stage 0, n = 5; I, n = 4; II, n = 15; III, n = 19; and IV, n = 33) and 98 patients with non-pancreatic cancer (intraductal papillary mucinous neoplasm, n = 36; chronic pancreatitis, n = 33; pancreatic neuroendocrine neoplasm, n = 8; autoimmune pancreatitis, n = 9; and others, n = 12) to evaluate diagnostic performance. Results: APOA2 showed lower accuracy for advanced (stages II-IV) pancreatic cancer compared to CA 19-9 (sensitivity, 50.7% vs. 83.6%; sensitivity, 77.6% vs. 87.9%), but it provided superior accuracy for early-stage (stages 0 and I) detection (sensitivity, 33.3% vs. 22.2%; specificity, 66.7% vs. 59.4%). Three early-stage pancreatic cancer cases negative for CA 19-9 were detected with the APOA2-i Index, demonstrating high diagnostic accuracy for early-stage pancreatic cancer when both biomarkers are combined (sensitivity, 44.4%; specificity, 46.7%). The multivariate analysis revealed pancreatic cancer to be an independent risk factor for APOA2-i Index positivity (odds ratio [OR]: 3.48, p < 0.001), CA 19-9 positivity (OR: 25.5, p < 0.001), and positivity for either marker (OR: 13.3, p < 0.001). Conclusions: The APOA2-i Index, combined with CA 19-9, may improve early-stage pancreatic cancer detection, especially in challenging cases and for high-risk patient surveillance.
Collapse
Affiliation(s)
- Kento Shionoya
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Atsushi Sofuni
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
- Department of Clinical Oncology, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Shuntaro Mukai
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Takayoshi Tsuchiya
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Reina Tanaka
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Ryosuke Tonozuka
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Kenjiro Yamamoto
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Kazumasa Nagai
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Yukitoshi Matsunami
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Hiroyuki Kojima
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Hirohito Minami
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Noriyuki Hirakawa
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Kyoko Asano
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Yuma Yamaguchi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Kazuki Hama
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| | - Takao Itoi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo 160-0023, Japan; (K.S.); (A.S.); (S.M.); (T.T.); (R.T.); (R.T.); (K.Y.); (K.N.); (Y.M.); (H.K.); (H.M.); (N.H.); (K.A.); (Y.Y.); (K.H.)
| |
Collapse
|
45
|
Wu X, Rong L, Tang R, Li Q, Wang F, Deng X, Miao L. Unveiling the role of CXCL10 in pancreatic cancer progression: A novel prognostic indicator. Open Med (Wars) 2025; 20:20241117. [PMID: 40129528 PMCID: PMC11931664 DOI: 10.1515/med-2024-1117] [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: 04/29/2024] [Revised: 11/14/2024] [Accepted: 11/25/2024] [Indexed: 03/26/2025] Open
Abstract
Objective Pancreatic cancer is distinguished by its high likelihood of metastasis and drug resistance, while the fundamental mechanisms are inadequately elucidated. This study aimed to identify pivotal hub genes associated with pancreatic cancer and assess their potential utility in predicting its onset and progression. Methods Weighted gene co-expression network analysis (WGCNA) combined with differential expression analysis identified novel susceptibility modules and hub genes for pancreatic cancer. Kyoto Encyclopedia of Genes and Genomes and gene ontology analyses were utilized to explore the potential roles of these hub genes. Receiver operator characteristic curves and nomogram models were developed to evaluate diagnostic efficacy. Mendelian randomization, flow cytometry, Transwell, and RNA sequencing were conducted to explore the association between C-X-C motif chemokine ligand 10 (CXCL10) and immune infiltration. Results WGCNA analysis was performed to build gene co-expression networks, and ten key genes were found. CXCL10 was the central gene, and its expression was significantly linked to the survival of patients with pancreatic cancer and their response to immune checkpoint inhibitors. CXCL10 demonstrated the ability to stimulate the differentiation of macrophages toward the M2 phenotype. CXCL10 could facilitate the metastasis of pancreatic cancer cells by modulating macrophage polarization. CXCL10 affects macrophage polarization by regulating the expression of vascular endothelial growth factor A. Conclusions CXCL10 demonstrates potential as a therapeutic target for managing pancreatic cancer.
Collapse
Affiliation(s)
- Xiaochao Wu
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Longfei Rong
- Department of General Surgery, SIR RUN RUN Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ruiyi Tang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Quanpeng Li
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fei Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xueting Deng
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lin Miao
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
46
|
Xiao Y, Sun S, Zheng N, Zhao J, Li X, Xu J, Li H, Du C, Zeng L, Zhang J, Yin X, Huang Y, Yang X, Yuan F, Jia X, Li B, Li B. Development of PDAC diagnosis and prognosis evaluation models based on machine learning. BMC Cancer 2025; 25:512. [PMID: 40114140 PMCID: PMC11924714 DOI: 10.1186/s12885-025-13929-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is difficult to detect early and highly aggressive, often leading to poor patient prognosis. Existing serum biomarkers like CA19-9 are limited in early diagnosis, failing to meet clinical needs. Machine learning (ML)/deep learning (DL) technologies have shown great potential in biomedicine. This study aims to establish PDAC differential diagnosis and prognosis assessment models using ML combined with serum biomarkers for early diagnosis, risk stratification, and personalized treatment recommendations, improving early diagnosis rates and patient survival. METHODS The study included serum biomarker data and prognosis information from 117 PDAC patients. ML models (Random Forest (RF), Neural Network (NNET), Support Vector Machine (SVM), and Gradient Boosting Machine (GBM)) were used for differential diagnosis, evaluated by accuracy, Kappa test, ROC curve, sensitivity, and specificity. COX proportional hazards model and DeepSurv DL model predicted survival risk, compared by C-index and Log-rank test. Based on DeepSurv's risk predictions, personalized treatment recommendations were made and their effectiveness assessed. RESULTS Effective PDAC diagnosis and prognosis models were built using ML. The validation set data shows that the accuracy of the RF, NNET, SVM, and GBM models are 84.21%, 84.21%, 76.97%, and 83.55%; the sensitivity are 91.26%, 90.29%, 89.32%, and 88.35%; and the specificity are 69.39%, 71.43%, 51.02%, and 73.47%. The Kappa values are 0.6266, 0.6307, 0.4336, and 0.6215; and the AUC are 0.889, 0.8488, 0.8488, and 0.8704, respectively. BCAT1, AMY, and CA12-5 were selected as modeling parameters for the prognosis model using COX regression. DeepSurv outperformed the COX model on both training and validation sets, with C-indexes of 0.738 and 0.724, respectively. The Kaplan-Meier survival curves indicate that personalized treatment recommendations based on DeepSurv can help patients achieve survival benefits. CONCLUSION This study built efficient PDAC diagnosis and prognosis models using ML, improving early diagnosis rates and prognosis accuracy. The DeepSurv model excelled in prognosis prediction and successfully guided personalized treatment recommendations and supporting PDAC clinical management.
Collapse
Affiliation(s)
- Yingqi Xiao
- Department of Clinical Laboratory, Beijing Electric Power Teaching Hospital, Capital Medical University, Beijing, China
| | - Shixin Sun
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Naxin Zheng
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Jing Zhao
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Xiaohan Li
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Jianmin Xu
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Haolian Li
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Chenran Du
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Lijun Zeng
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Juling Zhang
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Xiuyun Yin
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Yuan Huang
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Xuemei Yang
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Fang Yuan
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Xingwang Jia
- Department of Clinical Laboratory, Beijing Electric Power Teaching Hospital, Capital Medical University, Beijing, China.
| | - Boan Li
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China.
| | - Bo Li
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
47
|
Vincenzi MM, Mori M, Passoni P, Tummineri R, Slim N, Midulla M, Palazzo G, Belardo A, Spezi E, Picchio M, Reni M, Chiti A, del Vecchio A, Fiorino C, Di Muzio NG. Temporal Validation of an FDG-PET-Radiomic Model for Distant-Relapse-Free-Survival After Radio-Chemotherapy for Pancreatic Adenocarcinoma. Cancers (Basel) 2025; 17:1036. [PMID: 40149369 PMCID: PMC11941493 DOI: 10.3390/cancers17061036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Pancreatic cancer is a very aggressive disease with a poor prognosis, even when diagnosed at an early stage. This study aimed to validate and refine a radiomic-based [18F]FDG-PET model to predict distant relapse-free survival (DRFS) in patients with unresectable locally advanced pancreatic cancer (LAPC). Methods: A Cox regression model incorporating two radiomic features (RFs) and cancer stage (III vs. IV) was temporally validated using a larger cohort (215 patients treated between 2005-2022). Patients received concurrent chemoradiotherapy with capecitabine and hypo-fractionated Intensity Modulated Radiotherapy (IMRT). Data were split into training (145 patients, 2005-2017) and validation (70 patients, 2017-2022) groups. Seventy-eight RFs were extracted, harmonized, and analyzed using machine learning to develop refined models. Results: The model incorporating Statistical-Percentile10, Morphological-ComShift, and stage demonstrated moderate predictive accuracy (training: C-index = 0.632; validation: C-index = 0.590). When simplified to include only Statistical-Percentile10, performance improved slightly in the validation group (C-index = 0.601). Adding GLSZM3D-grayLevelVariance to Statistical-Percentile10, while excluding Morphological-ComShift, further enhanced accuracy (training: C-index = 0.654; validation: C-index = 0.623). Despite these refinements, all versions showed similar moderate ability to stratify patients into risk classes. Conclusions: [18F]FDG-PET radiomic features are robust predictors of DRFS after chemoradiotherapy in LAPC. Despite moderate performance, these models hold promise for patient risk stratification. Further validation with external cohorts is ongoing.
Collapse
Affiliation(s)
- Monica Maria Vincenzi
- Medical Physics, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.M.V.)
| | - Martina Mori
- Medical Physics, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.M.V.)
| | - Paolo Passoni
- Radiotherapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Roberta Tummineri
- Radiotherapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Najla Slim
- Radiotherapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Martina Midulla
- Radiotherapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gabriele Palazzo
- Medical Physics, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.M.V.)
| | - Alfonso Belardo
- Medical Physics, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.M.V.)
| | - Emiliano Spezi
- School of Engineering, Cardiff University, Cardiff CF24 4HQ, UK
| | - Maria Picchio
- Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Department of Medical Oncology, Faculty of Medicine and Surgery, Vita-Salute University, 20132 Milan, Italy
| | - Michele Reni
- Department of Medical Oncology, Faculty of Medicine and Surgery, Vita-Salute University, 20132 Milan, Italy
- Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Arturo Chiti
- Nuclear Medicine, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Department of Medical Oncology, Faculty of Medicine and Surgery, Vita-Salute University, 20132 Milan, Italy
| | - Antonella del Vecchio
- Medical Physics, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.M.V.)
| | - Claudio Fiorino
- Medical Physics, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (M.M.V.)
| | - Nadia Gisella Di Muzio
- Radiotherapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Department of Imaging Diagnostics, Neuroradiology, and Radiotherapy, Faculty of Medicine and Surgery, Vita-Salute University, 20132 Milan, Italy
| |
Collapse
|
48
|
Wu Y, Li T, Zhang R, Shi T, Wang S, Zhu L, Zhang Y, Zheng X, Yu X, Zhang J. Establishment of nomogram of early death in elderly pancreatic cancer patients with liver metastasis. Discov Oncol 2025; 16:333. [PMID: 40095230 PMCID: PMC11914455 DOI: 10.1007/s12672-025-02059-4] [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: 10/12/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Many elderly patients with pancreatic cancer (PC) often have liver metastasis (LM), and these patients often have poor prognosis and early death (ED). However, few models can accurately predict ED from elderly PC patients with LM. Therefore, we aim to create nomograms to predict ED in elderly PC patients with LM. METHODS All elderly (≥ 60 years old) PC patients with LM from 2010 to 2020 were downloaded from the Surveillance, Epidemiology, and End Result (SEER) database according to the admission criteria. The included data was randomly divided into the training set and the validation set, with a ratio of 7:3. The risk factors for ED in elderly PC patients with LM were determined by univariate and multivariate logistic regression methods, and a nomogram model was established. Lastly, the nomogram is verified by the receiver operating characteristic (ROC) curve, area under the curve (AUC), calibration curve, and decision curve analysis (DCA). RESULTS A total of 1,424 elderly PC patients with LM were randomly divided into training set (n = 996) and validation set (n = 428) based on the ratio of 7:3. The independent prognostic factors for ED include T stage, N stage, surgery, chemotherapy, lung metastasis, and other metastases. These variables were used to create nomograms, where the AUC of the training set and the validation set were 0.83 (95% CI 0.80-0.85) and 0.81 (95% CI 0.77-0.85), respectively. Furthermore, the calibration curve shows that the predicted ED is in good agreement with the actual value. DCA also shows good clinical application value. CONCLUSIONS The developed nomogram can be used to predict the specific probability of ED in elderly PC patients with LM, which is useful in guiding the early prevention and treatment decision-making of this group of people.
Collapse
Affiliation(s)
- Yang Wu
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Tian Li
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
- Department of Nephrology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
| | - Runbing Zhang
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Tingting Shi
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Shunna Wang
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Lingling Zhu
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Yani Zhang
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Xiaofeng Zheng
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Xiaohui Yu
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
| | - Jiucong Zhang
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China.
| |
Collapse
|
49
|
Turlej E, Domaradzka A, Radzka J, Drulis-Fajdasz D, Kulbacka J, Gizak A. Cross-Talk Between Cancer and Its Cellular Environment-A Role in Cancer Progression. Cells 2025; 14:403. [PMID: 40136652 PMCID: PMC11940884 DOI: 10.3390/cells14060403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
The tumor microenvironment is a dynamic and complex three-dimensional network comprising the extracellular matrix and diverse non-cancerous cells, including fibroblasts, adipocytes, endothelial cells and various immune cells (lymphocytes T and B, NK cells, dendritic cells, monocytes/macrophages, myeloid-derived suppressor cells, and innate lymphoid cells). A constantly and rapidly growing number of studies highlight the critical role of these cells in shaping cancer survival, metastatic potential and therapy resistance. This review provides a synthesis of current knowledge on the modulating role of the cellular microenvironment in cancer progression and response to treatment.
Collapse
Affiliation(s)
- Eliza Turlej
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Aleksandra Domaradzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Justyna Radzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Dominika Drulis-Fajdasz
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Julita Kulbacka
- Departament of Molecular and Cellular Biology, Faculty of Pharmacy, Wrocław Medical University, Borowska 211A, 50-556 Wrocław, Poland;
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Agnieszka Gizak
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| |
Collapse
|
50
|
Zhang S, Zhang Y, Feng S, Han M, Wang Z, Qiao D, Tian J, Wang L, Du B, Zhang Z, Zhong J. Tumor-promoting effect and tumor immunity of SRSFs. Front Cell Dev Biol 2025; 13:1527309. [PMID: 40129567 PMCID: PMC11931056 DOI: 10.3389/fcell.2025.1527309] [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: 11/13/2024] [Accepted: 02/17/2025] [Indexed: 03/26/2025] Open
Abstract
Serine/arginine-rich splicing factors (SRSFs) are a family of 12 RNA-binding proteins crucial for the precursor messenger RNA (pre-mRNA) splicing. SRSFs are involved in RNA metabolism events such as transcription, translation, and nonsense decay during the shuttle between the nucleus and cytoplasm, which are important components of genome diversity and cell viability. SRs recognize splicing elements on pre-mRNA and recruit the spliceosome to regulate splicing. In tumors, aberrant expression of SRSFs leads to aberrant splicing of RNA, affecting the proliferation, migration, and anti-apoptotic ability of tumor cells, highlighting the therapeutic potential of targeted SRSFs for the treatment of diseases. The body's immune system is closely related to the occurrence and development of tumor, and SRSFs can affect the function of immune cells in the tumor microenvironment by regulating the alternative splicing of tumor immune-related genes. We review the important role of SRSFs-induced aberrant gene expression in a variety of tumors and the immune system, and prospect the application of SRSFs in tumor. We hope that this review will inform future treatment of the disease.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yongxi Zhang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Sijia Feng
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Miaomiao Han
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Zixi Wang
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Dan Qiao
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jiaqi Tian
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Lan Wang
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Baoshun Du
- Second Department of Neurosurgery, Xinxiang Central Hospital, Xinxiang, China
| | - Zheying Zhang
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jiateng Zhong
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Henan Province Engineering Technology Research Center of Tumor diagnostic biomarkers and RNA interference drugs, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|