1
|
Lo Presti E, Cupaioli F, Scimeca D, Unti E, Di Martino V, Daidone R, Amata M, Scibetta N, Soucie E, Meraviglia S, Iovanna J, Dusetti N, De Gaetano A, Merelli I, Di Mitri R. The pancreatic tumor microenvironment of treatment-naïve patients causes a functional shift in γδ T cells, impairing their anti-tumoral defense. Oncoimmunology 2025; 14:2466301. [PMID: 39945298 PMCID: PMC11834455 DOI: 10.1080/2162402x.2025.2466301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 01/14/2025] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents a unique challenge for researchers due to its late diagnosis caused by vague symptoms and lack of early detection markers. Additionally, PDAC is characterized by an immunosuppressive microenvironment (TME), making it a difficult tumor to treat. While γδ T cells have shown potential for anti-tumor activity, conflicting studies exist regarding their effectiveness in pancreatic cancer. This study aims to explore the hypothesis that the PDAC TME hinders the anti-tumor capabilities of γδ T cells through blockade of cytotoxic functions. For this reason, we chose to enroll PDAC treatment-naive patients to avoid the possibility of therapy modifying the TME. By flow cytometry, our research findings indicate that the presence of γδ T cells among CD45+ cells in tumor tissue is lower compared to CD66+ cells, but higher than in blood. Circulating Vδ1 T cells exhibit a terminal effector memory phenotype (TEMRA) more than Vδ2 T cells. Interestingly, Vδ1 and Vδ2 T cells appear to be more prevalent at different stages of tumor development. In our in vitro culture using conditioned medium derived from Patient-derived organoids ;(PDOs), we observed a shift in expression markers in γδ T cells of healthy individuals toward an activation and exhaustion phenotype, as confirmed by scRNA-seq analysis extracted from a public database. A deeper understanding of γδ T cells in PDAC could be valuable for developing novel therapies aimed at mitigating the impact of the pancreatic tumor microenvironment on this cell population.
Collapse
Affiliation(s)
- Elena Lo Presti
- National Research Council of Italy (CNR), Institute for Biomedical Research and Innovation (IRIB), Palermo, Italy
| | - Francesca Cupaioli
- National Research Council of Italy, Bioinformatics Research Unit, Institute for Biomedical Technologies Segrate, Milan, Italy
| | - Daniela Scimeca
- Gastroenterology and Endoscopy Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| | - Elettra Unti
- ‘Anatomic-pathology Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| | - Vincenzo Di Martino
- Immunohaematology and Transfusion Medicine Unit, Imperia Hospital ASL1 Imperiese, Imperia, Italy
| | - Rossella Daidone
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Marseille, France
| | - Michele Amata
- Gastroenterology and Endoscopy Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| | - Nunzia Scibetta
- ‘Anatomic-pathology Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| | - Erinn Soucie
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Marseille, France
| | - Serena Meraviglia
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy
| | - Juan Iovanna
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Marseille, France
| | - Nelson Dusetti
- Cancer Research Center of Marseille (CRCM), INSERM, CNRS, Aix-Marseille University, Marseille, France
| | - Andrea De Gaetano
- National Research Council of Italy (CNR), Institute for Biomedical Research and Innovation (IRIB), Palermo, Italy
- National Research Council of Italy, Institute for Systems Analysis and Computer Science “A. Ruberti, ” BioMatLab (Biomathematics Laboratory), Rome, Italy
- Department of Mathematics, Mahidol University, Bangkok, Thailand
| | - Ivan Merelli
- National Research Council of Italy, Bioinformatics Research Unit, Institute for Biomedical Technologies Segrate, Milan, Italy
| | - Roberto Di Mitri
- Gastroenterology and Endoscopy Unit, Arnas Civico Di Cristina Benfratelli Hospital, Palermo, Italy
| |
Collapse
|
2
|
Victor Atoki A, Aja PM, Shinkafi TS, Ondari EN, Adeniyi AI, Fasogbon IV, Dangana RS, Shehu UU, Akin-Adewumi A. Exploring the versatility of Drosophila melanogaster as a model organism in biomedical research: a comprehensive review. Fly (Austin) 2025; 19:2420453. [PMID: 39722550 DOI: 10.1080/19336934.2024.2420453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 12/28/2024] Open
Abstract
Drosophila melanogaster is a highly versatile model organism that has profoundly advanced our understanding of human diseases. With more than 60% of its genes having human homologs, Drosophila provides an invaluable system for modelling a wide range of pathologies, including neurodegenerative disorders, cancer, metabolic diseases, as well as cardiac and muscular conditions. This review highlights key developments in utilizing Drosophila for disease modelling, emphasizing the genetic tools that have transformed research in this field. Technologies such as the GAL4/UAS system, RNA interference (RNAi) and CRISPR-Cas9 have enabled precise genetic manipulation, with CRISPR-Cas9 allowing for the introduction of human disease mutations into orthologous Drosophila genes. These approaches have yielded critical insights into disease mechanisms, identified novel therapeutic targets and facilitated both drug screening and toxicological studies. Articles were selected based on their relevance, impact and contribution to the field, with a particular focus on studies offering innovative perspectives on disease mechanisms or therapeutic strategies. Our findings emphasize the central role of Drosophila in studying complex human diseases, underscoring its genetic similarities to humans and its effectiveness in modelling conditions such as Alzheimer's disease, Parkinson's disease and cancer. This review reaffirms Drosophila's critical role as a model organism, highlighting its potential to drive future research and therapeutic advancements.
Collapse
Affiliation(s)
| | - Patrick Maduabuchi Aja
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- Department of Biochemistry, Faculty of Science, Ebonyi State University, Abakaliki, Nigeria
| | | | - Erick Nyakundi Ondari
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- School of Pure and Applied Sciences, Department of Biological Sciences, Kisii University, Kisii, Kenya
| | | | | | | | - Umar Uthman Shehu
- Department of Physiology, Kampala International University, Ishaka, Uganda
| | | |
Collapse
|
3
|
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; 122:678-689. [PMID: 40185208 DOI: 10.1016/j.ijrobp.2025.03.039] [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: 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
|
4
|
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
|
5
|
Sato-Dahlman M, Miura Y, Hajeri P, Roach B, Jacobsen K, Yamamoto M. Systemic therapy with the infectivity-selective oncolytic adenovirus by targeting mesothelin. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200967. [PMID: 40226846 PMCID: PMC11987630 DOI: 10.1016/j.omton.2025.200967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025]
Abstract
Treatment of advanced stage cancers is extremely challenging, and more effective systemic therapy is needed. Oncolytic adenoviruses (OAds) are one of the most promising anti-cancer agents. However, systemic delivery of OAd is challenging due to the low transduction in tumor cells caused by non-selective distribution and sequestration by non-target organs. To overcome this issue, we have previously generated a mesothelin (MSLN)-targeted OAd (AdML-VTIN). Here, we are reporting the potential of MSLN-targeted OAd as an agent for novel systemic treatment using MSLN-expressing lung and pancreatic cancer models. The in vivo biodistribution of AdML-VTIN after intravenous injection showed significantly lower liver sequestration compared to the wild type of OAd (AdML-5WT). By day 7, the intratumoral viral copy number of AdML-VTIN was significantly higher than that of AdML-5WT. For therapeutic efficacy, systemically injected AdML-VTIN exhibited statistically significant anti-tumor effects in both lung and pancreatic cancer xenograft tumor models. In addition, we tested the effect of preexisting immunity using human serum. In a neutralization assay, AdML-VTIN was more resistant to preexisting antibodies, compared to Ad5-WT. Interestingly, the hemagglutination profile of AdML-VTIN was also changed. Our results indicate that MSLN-targeted OAd has great potential to facilitate systemic therapy of advanced cancers.
Collapse
Affiliation(s)
- Mizuho Sato-Dahlman
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Yoshiaki Miura
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | | | - Brett Roach
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Kari Jacobsen
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
6
|
Alpsoy A, Yavuz A, Simsek K, Altunay B, Karaca M, Unal B, Bassorgun CI, Tatli AM, Elpek GO. Evaluation of tumor budding, desmoplastic reaction, and lymphocytic infiltration in predicting survival for pancreatic ductal adenocarcinoma. World J Gastrointest Oncol 2025; 17:107021. [DOI: 10.4251/wjgo.v17.i6.107021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/15/2025] [Accepted: 05/19/2025] [Indexed: 06/13/2025] Open
Abstract
BACKGROUND Although previous findings indicated that pathological assessment of tumor budding (TB), desmoplastic reaction (DR), and tumor-infiltrating lymphocytes (TILs) may play a role in determining tumor behavior in many malignancies, the relationship between TB, DR, and TILs in patients with pancreatic ductal adenocarcinoma (PDAC) is still unknown.
AIM To evaluate relationships of TB, DR, and TILs with histopathological parameters and determine their prognostic value in patients with PDAC.
METHODS The study cohort comprised 100 patients diagnosed with PDAC. Peritumoral budding (PTB) and intratumoral budding (ITB) were assessed according to the International Tumor Budding Consensus Conference guidelines. DR was classified based on stromal maturation. TILs were evaluated semiquantitatively with a 5% cutoff. Additionally, cases were categorized into two groups according to lymphocyte density: No/Low lymphocytes and medium/high lymphocytes.
RESULTS A significant correlation was observed between ITB and PTB (r = 0.890). Higher PTB was associated with fewer TILs and immature stroma (P < 0.001). PTB and TILs were significantly related to tumor dimension, lymphovascular invasion, lymph node metastasis (LNM), and stage (P < 0.005). ITB was also associated with the presence of lymph node involvement. The results of the univariate analysis revealed a significant correlation between poor survival rates and the presence of lymphovascular invasion, LNM, PTB, ITB, and TILs according to scoring (P < 0.001). The multivariate analysis revealed LNM, PTB, ITB, and TILs according to scoring as independent prognostic factors.
CONCLUSION TB assessment stratified patients with PDAC. PTB-ITB correlation showed diagnostic relevance of ITB in biopsy specimens. The prognostic significance of DR and interplay with TIL subsets warrant further investigation.
Collapse
Affiliation(s)
- Anıl Alpsoy
- Afyonkarahisar State Hospital, Afyonkarahisar 03000, Türkiye
| | - Aysen Yavuz
- Department of Pathology, Akdeniz University, School of Medicine, Antalya 07070, Türkiye
| | - Kubra Simsek
- Department of Pathology, Akdeniz University, School of Medicine, Antalya 07070, Türkiye
| | | | - Mustafa Karaca
- Department of Internal Medicine, Akdeniz University, Antalya 07070, Türkiye
| | - Betul Unal
- Antalya Bilim University, Antalya 07070, Türkiye
| | - Cumhur I Bassorgun
- Department of Pathology, Akdeniz University, School of Medicine, Antalya 07070, Türkiye
| | - Ali M Tatli
- Department of Oncology, Akdeniz University, School of Medicine, Antalya 07070, Türkiye
| | - Gulsum O Elpek
- Department of Pathology, Akdeniz University, School of Medicine, Antalya 07070, Türkiye
| |
Collapse
|
7
|
Bigot K, Patinote C, Garambois V, Chouchou A, Gayraud-Paniagua S, Vie N, Maggipinto Y, Smyej E, Robin M, Machu M, Bruciamacchie M, Colombo PE, Bousquet C, Mathonnet M, Levy-Augé E, Tosi D, Bonnet PA, Gongora C, Deleuze-Masquéfa C, Larbouret C. Inhibiting microtubule polymerization with EAPB02303, a prodrug activated by catechol-O-methyl transferase, enhances paclitaxel effect in pancreatic cancer models. Cell Death Dis 2025; 16:441. [PMID: 40490448 DOI: 10.1038/s41419-025-07747-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/30/2025] [Accepted: 05/20/2025] [Indexed: 06/11/2025]
Abstract
The Imiqualines family is an original group of small heterocyclic compounds, diversely substituted around different scaffolds. Among these compounds, the lead EAPB02303 displays outstanding cytotoxic activity at nanomolar concentrations comparable to those of standard-of-care chemotherapy drugs in different cancer cell lines, including Pancreatic Ductal AdenoCarcinoma (PDAC) cell lines. Due to its high aggressiveness and resistance to therapies, PDAC has an extremely poor prognosis with limited treatment options. Here, we demonstrated the cytotoxic activities of EAPB02303 alone or combined with standard chemotherapy drugs in several PDAC cell lines and confirmed these results in patient-derived xenograft mouse models. EAPB02303 potently induced cell cycle arrest in the G2/M phase and in mitosis followed by apoptosis. Then, using a combination of transcriptomic, proteomic, biochemical and cellular assay, we found that EAPB02303 mechanism of action relies on its bioactivation by catechol-O-methyltransferase, resulting in the production of a methylated compound that effectively inhibits microtubule polymerization. Moreover, EAPB02303 had a synergistic effect when combined with paclitaxel (the standard-of-care agent in PDAC) providing the rationale to continue the development of EAPB02303 combination strategies for the treatment of catechol-O-methyltransferase-overexpressing PDAC.
Collapse
Affiliation(s)
- Kévin Bigot
- IRCM, Université de Montpellier, Inserm, ICM, Montpellier, France
| | - Cindy Patinote
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, (CNRS, ENSCM, Université de Montpellier), Montpellier, France
| | | | - Adrien Chouchou
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, (CNRS, ENSCM, Université de Montpellier), Montpellier, France
| | - Stéphanie Gayraud-Paniagua
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, (CNRS, ENSCM, Université de Montpellier), Montpellier, France
| | - Nadia Vie
- IRCM, Université de Montpellier, Inserm, ICM, Montpellier, France
| | - Yann Maggipinto
- IRCM, Université de Montpellier, Inserm, ICM, Montpellier, France
| | - Elias Smyej
- IRCM, Université de Montpellier, Inserm, ICM, Montpellier, France
| | - Mathilde Robin
- IRCM, Université de Montpellier, Inserm, ICM, Montpellier, France
| | - Margot Machu
- IRCM, Université de Montpellier, Inserm, ICM, Montpellier, France
| | | | | | - Corinne Bousquet
- Université Toulouse III-Paul Sabatier - Centre de Recherche en Cancérologie de Toulouse (CRCT) - UMR1037 Inserm- UMR, 5071 CNRS, Toulouse, France
| | | | - Ela Levy-Augé
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, (CNRS, ENSCM, Université de Montpellier), Montpellier, France
| | - Diego Tosi
- IRCM, Université de Montpellier, Inserm, ICM, Montpellier, France
| | - Pierre-Antoine Bonnet
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, (CNRS, ENSCM, Université de Montpellier), Montpellier, France
| | - Céline Gongora
- IRCM, Université de Montpellier, Inserm, ICM, CNRS, Montpellier, France
| | - Carine Deleuze-Masquéfa
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, (CNRS, ENSCM, Université de Montpellier), Montpellier, France.
| | | |
Collapse
|
8
|
Madar S, Amor RE, Furman-Assaf S, Friedman E. Innovative Approaches to Early Detection of Cancer-Transforming Screening for Breast, Lung, and Hard-to-Screen Cancers. Cancers (Basel) 2025; 17:1867. [PMID: 40507348 PMCID: PMC12153683 DOI: 10.3390/cancers17111867] [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: 03/14/2025] [Revised: 05/25/2025] [Accepted: 05/29/2025] [Indexed: 06/16/2025] Open
Abstract
Early detection of cancer is crucial for improving patient outcomes. Traditional modalities such as mammography and low-dose computed tomography are effective but exhibit inherent limitations, including radiation exposure and accessibility challenges. This review explores innovative, non-invasive cancer screening methods, focusing on liquid biopsy and volatile organic compound (VOC)-based detection platforms. Liquid biopsy analyzes circulating tumor DNA and other biomarkers in bodily fluids, offering potential for early detection and monitoring of treatment response. VOC-based detection leverages unique metabolic signatures emitted by cancer cells, detectable in exhaled breath or other bodily emissions, providing a rapid and patient-friendly screening option. We provide a comprehensive overview of these advanced multi-cancer detection techniques to enhance diagnostic accuracy, accessibility, and patient adherence, and ultimately enhance survival rates and patient outcomes.
Collapse
Affiliation(s)
| | | | | | - Eitan Friedman
- Assuta Medical Centers, Tel-Aviv 6971028, Israel;
- Gray Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| |
Collapse
|
9
|
Fang Y, Tan C, Zheng Z, Yang J, Tang J, Guo R, Silli EK, Chen Z, Chen J, Ge R, Liu Y, Wen X, Liang J, Zhu Y, Jin Y, Li Q, Wang Y. The function of microRNA related to cancer-associated fibroblasts in pancreatic ductal adenocarcinoma. Biochem Pharmacol 2025; 236:116849. [PMID: 40056941 DOI: 10.1016/j.bcp.2025.116849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignant tumor characterized by a poor prognosis. A prominent feature of PDAC is the rich and dense stroma present in the tumor microenvironment (TME), which significantly hinders drug penetration. Cancer-associated fibroblasts (CAFs), activated fibroblasts originating from various cell sources, including pancreatic stellate cells (PSCs) and mesenchymal stem cells (MSCs), play a critical role in PDAC progression and TME formation. MicroRNAs (miRNAs) are small, single-stranded non-coding RNA molecules that are frequently involved in tumorigenesis and progression, exhibiting either oncolytic or oncogenic activity. Increasing evidence suggests that aberrant expression of miRNAs can mediate interactions between cancer cells and CAFs, thereby providing novel therapeutic targets for PDAC treatment. In this review, we will focus on the potential roles of miRNAs that target CAFs or CAFs-derived exosomes in PDAC progression, highlighting the feasibility of therapeutic strategies aimed at restoring aberrantly expressed miRNAs associated with CAFs, offering new pathways for the clinical management of PDAC.
Collapse
Affiliation(s)
- Yaohui Fang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Chunlu Tan
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenjiang Zheng
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianchen Yang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jiali Tang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruizhe Guo
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Epiphane K Silli
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Zhe Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jia Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruyu Ge
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yuquan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xiuqi Wen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jingdan Liang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yunfei Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yutong Jin
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Qian Li
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ying Wang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| |
Collapse
|
10
|
Dreyer SB, Beer P, Hingorani SR, Biankin AV. Improving outcomes of patients with pancreatic cancer. Nat Rev Clin Oncol 2025; 22:439-456. [PMID: 40329051 DOI: 10.1038/s41571-025-01019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 05/08/2025]
Abstract
Research studies aimed at improving the outcomes of patients with pancreatic ductal adenocarcinoma (PDAC) have brought about limited progress, and in clinical practice, the optimized use of surgery, chemotherapy and supportive care have led to modest improvements in survival that have probably reached a plateau. As a result, PDAC is expected to be the second leading cause of cancer-related death in Western societies within a decade. The development of therapeutic advances in PDAC has been challenging owing to a lack of actionable molecular targets, a typically immunosuppressive microenvironment, and a disease course characterized by rapid progression and clinical deterioration. Yet, the progress in our understanding of PDAC and identification of novel therapeutic opportunities over the past few years is leading to a strong sense of optimism in the field. In this Perspective, we address the aforementioned challenges, including biological aspects of PDAC that make this malignancy particularly difficult to treat. We explore specific areas with potential for therapeutic advances, including targeting mutant KRAS, novel strategies to harness the antitumour immune response and approaches to early detection, and propose mechanisms to improve clinical trial design and to overcome various community and institutional barriers to progress.
Collapse
Affiliation(s)
- Stephan B Dreyer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- Department of Hepatobiliary Surgery, Royal Liverpool University Hospital, Liverpool, UK
| | - Philip Beer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- Hull York Medical School, University of York, York, UK
| | - Sunil R Hingorani
- Department of Internal Medicine, Division of Hemotology/Oncology, University of Nebraska Medical Center, Omaha, NE, USA
- Pancreatic Cancer Center of Excellence, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK.
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK.
| |
Collapse
|
11
|
Wang C, Song W, Zhang Y, Deng H, Zhou Z, Zhu J, Wang X. SKIL Promotes Pancreatic Cancer Metastasis by Inhibiting TSPYL2 to Activate the TGF-β Pathway. CANCER INNOVATION 2025; 4:e70011. [PMID: 40391200 PMCID: PMC12086372 DOI: 10.1002/cai2.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/24/2025] [Accepted: 04/07/2025] [Indexed: 05/21/2025]
Abstract
Background Pancreatic adenocarcinoma (PAAD) represents a highly fatal form of cancer. The 5-year survival rate for patients with this disease is only around 10%. A significant hurdle in its management is the absence of characteristic early-stage symptoms. As a result, a large majority of pancreatic cancer patients are diagnosed when the disease has reached an advanced stage or has metastasized. Consequently, taking measures to suppress the occurrence of metastasis in pancreatic cancer can bring about a substantial improvement in patients' survival rates and overall prognosis. SKIL, known to promote cancer progression, is implicated in cell proliferation, epithelial-mesenchymal transition (EMT), and metastasis, but its specific function in pancreatic cancer remains unclear. Methods We investigated the effects of SKIL on the proliferation, apoptosis, and metastasis of pancreatic cancer cells. Through ChIP-seq, we identified the SKIL downstream target gene and further explored the mechanism by which SKIL regulates the metastasis of pancreatic cancer cells through functional experiments and Western blot. Results A high level of SKIL expression is associated with an unfavorable prognosis in PAAD; it promotes cell migration and EMT. Through ChIP-seq analysis, we identified that SKIL inhibits TSPYL2, a nuclear protein regulating the TGF-β pathway by binding to the TGFB1 promoter. Further studies carried out by us confirmed that SKIL modulates the TGF-β pathway via TSPYL2, facilitating EMT and metastasis in pancreatic cancer cells, independent of Smad4. Conclusions These findings reveal a novel regulatory mechanism involving SKIL, TSPYL2, and the TGF-β pathway, offering new therapeutic targets for PAAD.
Collapse
Affiliation(s)
- Chenxi Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Weiwei Song
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yixuan Zhang
- NMPA Key Laboratory for Monitoring and Evaluation of Cosmetics, Shanghai Innovation R&DTesting and Evaluation Technical Service Platform of Cosmetics(22DZ2292100)ShanghaiChina
| | - Hongming Deng
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zixiang Zhou
- Department of Molecular BiologyPrinceton UniversityPrincetonNew JerseyUSA
| | - Jing Zhu
- College of Nursing and Health InnovationThe University of Texas ArlingtonArlingtonTexasUSA
| | - Xiaobing Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
12
|
Li BR, Wang T, Hu HF, Wu D, Zhou CJ, Ji SR, Zhuo QF, Li Z, Wang ZL, Fan GX, Jing DS, Yu CY, Qin Y, Chen XM, Xu JF, Xu XW. Acyl-CoA thioesterase 8 induces gemcitabine resistance via regulation of lipid metabolism and antiferroptotic activity in pancreatic ductal adenocarcinoma. Acta Pharmacol Sin 2025; 46:1742-1756. [PMID: 39939803 PMCID: PMC12098905 DOI: 10.1038/s41401-025-01477-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/08/2025] [Indexed: 02/14/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) comprises a group of highly malignant tumors of the pancreas. Metabolic reprogramming in tumors plays a pivotal role in promoting cancer progression. However, little is known about the metabolic alterations in tumors that drive cancer drug resistance in patients with PDAC. Here, we identified acyl-CoA thioesterase 8 (ACOT8) as a key player in driving PDAC gemcitabine (GEM) resistance. The expression of ACOT8 is significantly upregulated in GEM-resistant PDAC tissues and is closely associated with poor survival in patients with PDAC. Gain- and loss-of-function studies have shown that ACOT8 drives PDAC GEM resistance both in vitro and in vivo. Mechanistically, ACOT8 regulates cellular cholesterol ester (CE) levels, decreases the levels of phosphatidylethanolamines (PEs) that bind to polyunsaturated fatty acids and promote peroxisome activation. The knockdown of ACOT8 promotes ferroptosis and increases the chemosensitivity of tumors to GEM by inducing ferroptosis-associated pathway activation in PDAC cell lines. The combination of orlistat, an ACOT8 inhibitor, and GEM significantly inhibited tumor growth in PDAC organoid and mouse models. This study reveals the biological importance of ACOT8 and provides a potential combination therapy for treating patients with advanced GEM-resistant PDAC.
Collapse
Affiliation(s)
- Bo-Rui Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Hepatopancreatobiliary Surgery, First College of Clinical Medical Science, Three Gorges University, Yichang, 443003, China
- People's Hospital of China Three Gorges University, Yichang, 443099, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Ting Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Hai-Feng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
- Department of General Surgery, First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Di Wu
- Department of Hepatopancreatobiliary, Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Chen-Jie Zhou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Shun-Rong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Qi-Feng Zhuo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zheng Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zhi-Liang Wang
- Department of Hepatopancreatobiliary, Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Gui-Xiong Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - De-Sheng Jing
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chong-Yuan Yu
- Department of Hepatopancreatobiliary, Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xue-Min Chen
- Department of Hepatopancreatobiliary, Third Affiliated Hospital of Soochow University, Changzhou, 213000, China.
| | - Jun-Feng Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xiao-Wu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
13
|
Zhao L, Chen G, Li D, Wang K, Schaefer M, Herr I, Yan B. Baicalein disrupts TGF-β-induced EMT in pancreatic cancer by FTO-dependent m6A demethylation of ZEB1. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119969. [PMID: 40262723 DOI: 10.1016/j.bbamcr.2025.119969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 04/08/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy associated with poor prognosis. Baicalein, a flavonoid extracted from the roots of Scutellaria baicalensis, traditionally used in Chinese medicine, has demonstrated potential in inhibiting cancer development and progression. However, its mechanism of action remains poorly understood, particularly regarding epigenetic gene regulation through m6A RNA methylation. In this study, three human PDAC cell lines and one nonmalignant cell line were employed. The effects of baicalein were examined using multiple assays, including RT-qPCR, MeRIP-qPCR, Western blotting, spheroid formation, RNA stability, and MTT, to evaluate cellular functions and m6A regulation. Baicalein significantly reduced cell viability, migration, invasion, and colony formation. It also downregulated FTO, an enzyme critical for m6A RNA demethylation. Knockdown of FTO replicated the effects of baicalein, underscoring its oncogenic role in PDAC. Bioinformatic analysis identified ZEB1-a key transcription factor in epithelial-to-mesenchymal transition-as an m6A-modified target regulated by FTO. Both baicalein treatment and FTO knockdown enhanced m6A modification and decreased ZEB1 mRNA stability, thereby suppressing stemness-related features. Rescue experiments further confirmed that baicalein disrupts the TGF-β/FTO/ZEB1 signaling axis, highlighting its therapeutic potential in PDAC. This study offers fundamental insights for the development of novel therapeutic strategies targeting PDAC.
Collapse
Affiliation(s)
- Lian Zhao
- Department of General, Visceral & Transplant Surgery, Section Surgical Research, University of Heidelberg, Im Neuenheimer Feld 365, Heidelberg 69120, Germany.
| | - Gong Chen
- Department of General, Visceral & Transplant Surgery, Section Surgical Research, University of Heidelberg, Im Neuenheimer Feld 365, Heidelberg 69120, Germany
| | - Dan Li
- Department of General, Visceral & Transplant Surgery, Section Surgical Research, University of Heidelberg, Im Neuenheimer Feld 365, Heidelberg 69120, Germany
| | - Kangtao Wang
- Department of General, Visceral & Transplant Surgery, Section Surgical Research, University of Heidelberg, Im Neuenheimer Feld 365, Heidelberg 69120, Germany
| | - Michael Schaefer
- Department of General, Visceral & Transplant Surgery, Section Surgical Research, University of Heidelberg, Im Neuenheimer Feld 365, Heidelberg 69120, Germany
| | - Ingrid Herr
- Department of General, Visceral & Transplant Surgery, Section Surgical Research, University of Heidelberg, Im Neuenheimer Feld 365, Heidelberg 69120, Germany
| | - Bin Yan
- Department of General, Visceral & Transplant Surgery, Section Surgical Research, University of Heidelberg, Im Neuenheimer Feld 365, Heidelberg 69120, Germany.
| |
Collapse
|
14
|
Li Z, Duan J, Liu Z, Li W, Mai Y, Fu H, Yuan G, Wang J. A triple-mode strategy on JQ1-loaded nanoplatform for superior antitumor therapy in pancreatic cancer. Mater Today Bio 2025; 32:101696. [PMID: 40225138 PMCID: PMC11986615 DOI: 10.1016/j.mtbio.2025.101696] [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: 11/28/2024] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Pancreatic cancer's dire prognosis urgently calls for innovative therapeutic strategies. JQ1, a bromodomain 4 inhibitor, exhibits potent anti-tumor activity in preclinical models but faces limitations due to rapid resistance development. Here, we developed a novel multifunctional nanoplatform, JQ1@MSN/FeTA-iRGD, which implemented a triple-mode strategy integrating apoptosis, ferroptosis, and immunogenic cell death for optimized treatment of pancreatic cancer. The particles could precisely target tumors in mice and achieve efficient release of JQ1 and Fe2+ through internalization in the acidic tumor environment. The nanoplatform amplified reactive oxygen species and mitochondrial damage to disrupt the redox homeostasis, thus synergistically escalating apoptosis and ferroptosis for the destruction of tumor cells, circumventing the rapid drug resistance associated with monotherapy. Meanwhile, dying cancer cells released damage-associated molecular patterns, which facilitated immunogenic cell death and triggered antitumor immune responses, guaranteeing the sustained efficacy of the treatment. Moreover, the system exhibited favorable biocompatibility, supporting its feasibility for clinical translation. Our results demonstrated that this novel strategy, combining apoptosis, ferroptosis, and immunogenic cell death, overcame the limitations of monotherapy with JQ1, providing a superior, targeted, and sustainable treatment option for pancreatic cancer.
Collapse
Affiliation(s)
- Zhiguo Li
- Guangzhou Key Laboratory of Medical Nanomaterials, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Jinxin Duan
- Guangzhou Key Laboratory of Medical Nanomaterials, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Zhiwen Liu
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Weifan Li
- Guangzhou Key Laboratory of Medical Nanomaterials, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yiyin Mai
- Guangzhou Key Laboratory of Medical Nanomaterials, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Hao Fu
- Guangzhou Key Laboratory of Medical Nanomaterials, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Guotao Yuan
- College of Chemistry and Environmental Engineering, Shenzhen University, 518060, China
- Department of Otolaryngology, Longgang E.N.T. Hospital & Shenzhen Key Laboratory of E.N.T., Shenzhen, 518116, China
| | - Jiawei Wang
- Guangzhou Key Laboratory of Medical Nanomaterials, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| |
Collapse
|
15
|
Yu X, Chen X, Chen W, Han X, Xie Q, Geng D, Guo G, Zhou L, Tang S, Chen J, Huang X, Zhong X. TGFβ2 Promotes the Construction of Fibrotic and Immunosuppressive Tumor Microenvironment in Pancreatic Adenocarcinoma: A Comprehensive Analysis. Mol Biotechnol 2025; 67:2562-2575. [PMID: 39044066 DOI: 10.1007/s12033-024-01219-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 06/10/2024] [Indexed: 07/25/2024]
Abstract
Pancreatic adenocarcinoma (PAAD) was characterized by dense fibrotic stroma and immunosuppressive tumor microenvironment (TME). TGFβ signaling pathways are highly activated in human cancers. However, the role of TGFβ2 in TME of PAAD remains to be elucidated. In this study, we showed that TGFβ2 was expressed at a relatively high level in PAAD tissues or cancer cells. Moreover, its high expression predicted unfavorable prognosis. In PAAD, gene set enrichment analysis showed that TGFβ2 correlated positively with leukocyte transendothelial migration, but negatively with aerobic metabolism, including oxidative phosphorylation. Results in Tumor and Immune System Interaction Database showed that TGFβ2 correlated with the infiltration of tumor-associated macrophages (TAMs), which could be attributed to that TGFβ2 promote CCL2 expression in PAAD. Moreover, correlation analysis showed that TGFβ2 could trigger cancer-associated fibroblasts (CAFs) activation in PAAD. The drug sensitivity analysis may indicate that patients with TGFβ2 high expression have higher sensitivity to chemotherapeutics, but the sensitivity to targeted drugs is still controversial. TGFβ2 could promote expansion of CAFs and infiltration of TAMs, thus participating in the construction of a fibrotic and immunosuppressive TME in PAAD. Targeting TGFβ2 could be a promising therapeutic approach, which needs to be elucidated by clinical and experimental evidences.
Collapse
Affiliation(s)
- Xiaofen Yu
- Department of Medical Oncology, Nanchang Third Hospital, Nanchang, 330000, Jiangxi, China
| | - Xuefen Chen
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Wanxian Chen
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Xiaosha Han
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Qihu Xie
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Deyi Geng
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Genghong Guo
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Linsa Zhou
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Shijie Tang
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China
| | - Jiasheng Chen
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China.
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China.
| | - Xin Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Department of Pancreatobiliary Surgery, Sun Yat-Sen University Cancer Center, GuangzhouGuangdong, 510060, China.
| | - Xiaoping Zhong
- Department of Plastic and Burns Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China.
- Plastic Surgery Research Institute, Ear Deformities Treatment Center and Cleft Lip and Palate Treatment Center of Shantou University Medical College, Shantou, China.
| |
Collapse
|
16
|
Zhan Y, Zhang K, Fan Y, Lin S, Wu J, Xu H. Lipids, lipid-lowering drug target genes and pancreatic cancer: a Mendelian randomization study. Int J Clin Pharm 2025; 47:747-754. [PMID: 39821006 DOI: 10.1007/s11096-025-01866-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/04/2025] [Indexed: 01/19/2025]
Abstract
BACKGROUND Pancreatic cancer (PC) is a malignant tumor with a low survival rate. Lipid modifiers show potential for PC therapy, but evidence is lacking. AIM This Mendelian randomization (MR) study aimed to explore the relationship between lipid traits, and lipid-lowering drug target genes with PC risk. METHOD Genetic instrumental variables associated with lipid traits and lipid-lowering drug target genes were used to perform MR analyses of PC risk. MR estimation was based on genome-wide association study data from two large sample sets, and the MR results were meta-analyzed to assess their impact on PC risk. To ensure the reliability of lipid-modifying drug targets, we conducted a Summary Data-based Mendelian Randomization (SMR) analysis. Additionally, a two-step MR analysis was employed to explore potential mediating effects. RESULTS In two independent datasets, HMG-CoA reductase (HMGCR) inhibition was statistically associated with a lower risk of PC (OR 0.50, [95% CI 0.25-1.00]; p = 0.0453). The results were further supported by SMR analysis, which showed a similar association (OR 0.51, [95% CI 0.28-0.96]; p = 0.0369). Mediation analysis revealed that 11.69% of the protective effect of HMGCR inhibitors on PC is mediated through lower BMI levels. No significant effect of lipid traits and the other eight lipid-lowering drug targets on PC risk was found. CONCLUSION This study suggests that HMGCR may be a potential drug target for the treatment or prevention of PC, providing important insights into the use of lipid-targeted drugs in PC therapy.
Collapse
Affiliation(s)
- Yuxuan Zhan
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China
| | - Kai Zhang
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China
| | - Yiqun Fan
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Siyi Lin
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China
| | - Jian Wu
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China
| | - Hongxia Xu
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China.
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
17
|
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
|
18
|
Chuong MD, Ashman J, Jethwa K, Kharofa J, Kim H, Koay E, Ludmir E, Miller E, Nelson B, Reyngold M, Sanford N, Chang D. Moving From the Background Toward the Spotlight: A Critical Review of Radiation Therapy for Locally Advanced Pancreas Cancer. Int J Radiat Oncol Biol Phys 2025; 122:294-312. [PMID: 40032056 DOI: 10.1016/j.ijrobp.2025.02.022] [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] [Revised: 01/31/2025] [Accepted: 02/16/2025] [Indexed: 03/05/2025]
Abstract
Radiation therapy (RT) for locally advanced pancreatic cancer (LAPC) continues to be controversial. Advances in both systemic therapy and RT techniques have changed the landscape of LAPC management in recent years. Clinical outcomes of ablative RT have been encouraging, and randomized clinical trials may clarify the role of RT for LAPC. We present a contemporary critical review of key aspects regarding optimal patient selection, radiation dose escalation techniques, novel radiosensitizers and radioprotectors, and treatment response assessment for LAPC.
Collapse
Affiliation(s)
- Michael D Chuong
- Department of Radiation Oncology, Miami Cancer Institute, Miami, Florida.
| | - Jonathan Ashman
- Department of Radiation Oncology, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Krishan Jethwa
- Department of Radiation Oncology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Jordan Kharofa
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Hyun Kim
- Department of Radiation Oncology, Washington University in St. Louis, Missouri.
| | - Eugene Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ethan Ludmir
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eric Miller
- Department of Radiation Oncology, Ohio State University, Columbus, Ohio
| | - Bailey Nelson
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Marsha Reyngold
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nina Sanford
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Daniel Chang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
19
|
Darian S, Malo JS, Lim JS, Buell JF, Osman H, Van Meter T, Jeyarajah DR. Yttrium-90 Radioembolization for Intrahepatic Cholangiocarcinoma: Non-University Tertiary Care Center Experience. Surg Innov 2025; 32:229-234. [PMID: 39882666 DOI: 10.1177/15533506251317283] [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: 01/31/2025]
Abstract
BackgroundIntrahepatic cholangiocarcinoma (ICC) presents a significant clinical challenge due to its high fatality rate and limited surgical candidacy. With only 30-40% of patients eligible for surgery upon diagnosis, alternative therapies are imperative. This study assesses the efficacy of Yttrium-90 (Y-90) radioembolization for unresectable ICC patients in a non-university tertiary care center (NUTCC).MethodsA retrospective analysis of 15 unresectable ICC patients treated with Y-90 radioembolization was conducted. Tumor response, survival, and adverse events were evaluated using RECIST criteria.Results60% of patients exhibited partial response, and 20% showed stable disease, with notable tumor size reduction and a median survival of 14 months. Minimal adverse effects were observed, indicating Y-90's favorable safety profile.ConclusionY-90 radioembolization shows potential in reducing tumor burden and enhancing survival rates with minimal adverse effects for unresectable ICC. Larger prospective studies are needed to confirm its efficacy and define its role in ICC treatment protocols.
Collapse
Affiliation(s)
- Sahar Darian
- Burnett School of Medicine at Texas Christian University, Fort Worth, TX, USA
| | - Juan S Malo
- Department of Surgery, Methodist Richardson Medical Center, Richardson, TX, USA
| | - Joseph S Lim
- Department of Surgery, Methodist Richardson Medical Center, Richardson, TX, USA
| | - Joseph F Buell
- Department of Surgery, Methodist Richardson Medical Center, Richardson, TX, USA
| | - Houssam Osman
- Department of Surgery, Methodist Richardson Medical Center, Richardson, TX, USA
| | | | - D Rohan Jeyarajah
- Burnett School of Medicine at Texas Christian University, Fort Worth, TX, USA
- Department of Surgery, Methodist Richardson Medical Center, Richardson, TX, USA
| |
Collapse
|
20
|
Shi H, Liu X, Xing C, Guo S, Zheng Y, Tan W, Ge Y, Xu J, Li Y, Song J. DNMT1-Induced Downregulation of CBX7 Inhibits ERK Phosphorylation and Promotes Pancreatic Ductal Adenocarcinoma Progression. FASEB J 2025; 39:e70571. [PMID: 40387566 PMCID: PMC12087528 DOI: 10.1096/fj.202402903r] [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: 11/19/2024] [Revised: 02/25/2025] [Accepted: 04/16/2025] [Indexed: 05/20/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancer types, characterized by an alarmingly low 5-year survival rate. DNA methylation has been implicated in the progression of various tumors, with DNA methyltransferase 1 (DNMT1) being the most extensively studied enzyme in this context. However, the expression patterns and underlying mechanisms of DNMT1 in PDAC remain poorly understood. The levels of DNMT1 and CBX7 in PDAC tissues and cells were determined by IHC and Western blot. ChIP and dual-luciferase reporter assays confirmed the interaction between DNMT1 and the CBX7 promoter. Cellular functions were evaluated through CCK-8, wound healing, and transwell assays. The expression of MAPK-related proteins was analyzed by Western blot. DNMT1 expression was upregulated in PDAC tissues and cell lines, whereas CBX7 expression was downregulated. Silencing DNMT1 inhibited cell proliferation, migration, and invasion in PDAC by modulating CBX7 expression. Moreover, DNMT1 methylates the CBX7 promoter region, leading to increased ERK phosphorylation, which subsequently drives tumorigenesis and metastasis in PDAC. DNMT1 promotes the malignant progression of PDAC through the CBX7/ERK pathway. Our study provides evidence for potential therapeutic targets for the comprehensive treatment of PDAC.
Collapse
Affiliation(s)
- Haowei Shi
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Xu Liu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Cheng Xing
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Shiqi Guo
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Yangyang Zheng
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Wendan Tan
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Yunpeng Ge
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Jingyong Xu
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Yao Li
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| | - Jinghai Song
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingP. R. China
| |
Collapse
|
21
|
Jeong JH, Shin D, Kim SY, Bae DJ, Sung YH, Koh EY, Kim J, Kim CJ, Park JS, Choi JK, Kim SC, Jun E. Spatial distribution and activation changes of T cells in pancreatic tumors according to KRAS mutation subtype. Cancer Lett 2025; 618:217641. [PMID: 40090570 DOI: 10.1016/j.canlet.2025.217641] [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: 12/03/2024] [Revised: 03/03/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
To enhance immunotherapy efficacy in pancreatic cancer, it is crucial to characterize its immune landscape and identify key factors driving immune alterations. To achieve this, we quantitatively analyzed the immune microenvironment using multiplex immunohistochemistry, assessing the spatial relationships between immune and tumor cells to correlate with patient survival rates and oncological factors. Additionally, through Whole Exome Sequencing analysis based on public data, we explored genetic mutations that could drive these compositions. Finally, we validated T cell (Tc) migration mechanisms using patient-derived tumor organoids with induced KRAS mutation subtypes. Through this approach, we obtained the following meaningful results. First, immune cells in pancreatic cancer are denser in stromal regions than near tumor cells, with higher Tc distribution linked to increased patient survival rates. Second, the distance between tumor and Tc was within 100 μm, with higher Tc density found within 15-30 μm of the tumor cells. Third, while increasing CAF levels correspond to higher Tc density, higher ECM density tends to decrease Tc presence. Fourth, compared to KRAS G12D, KRAS G12V mutation increases various immune cells, notably Tc, which is closely linked to a dramatic rise in vascular cells. Finally, Tc migration was enhanced in tumor organoids with the G12V mutation, attributed to a reduction in the secretion of immunosuppressive cytokines. Our results indicate that KRAS mutation subtypes influence immune cell composition and function in the pancreatic cancer microenvironment, leading to varied immunotherapy responses. This underscores the need for personalized immune therapeutics and research models specific to KRAS mutations.
Collapse
Affiliation(s)
- Ji Hye Jeong
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Dakyum Shin
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation Surgery, Department of General Surgery, Chosun University Hospital, 365, Pilmun-daero, Dong-gu, Gwangju Metropolitan City, 61453, Republic of Korea
| | - Sang-Yeob Kim
- Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Dong-Jun Bae
- PrismCDX, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Young Hoon Sung
- Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Cell and Genetic Engineering, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Eun-Young Koh
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jinju Kim
- Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Chong Jai Kim
- Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jae Soon Park
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea; SCL-KAIST Institute of Translational Research, Daejeon, 34141, Republic of Korea
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea; SCL-KAIST Institute of Translational Research, Daejeon, 34141, Republic of Korea.
| | - Song Cheol Kim
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Surgery, BK21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| | - Eunsung Jun
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Department of Convergence Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea; Asan Preclinical Evaluation Center for Cancer Therapeutix, Asan Medical Center, Seoul, 05505, Republic of Korea; Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea.
| |
Collapse
|
22
|
Kazlow E, Rinawi E, Gerszman E, Mattar S, Essami N, Nasir M, Abu Shtaya A, Assaf W, Haddad R, Mahamid A. Higher Neutrophil-to-Lymphocyte Ratio and Systemic Immune-Inflammation Index Is Associated with Better Prognosis Following Pancreaticoduodenectomy for Pancreatic Adenocarcinoma. J Clin Med 2025; 14:3762. [PMID: 40507524 PMCID: PMC12155792 DOI: 10.3390/jcm14113762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/16/2025] [Accepted: 05/26/2025] [Indexed: 06/16/2025] Open
Abstract
Background: Pancreatic cancer has a high mortality rate worldwide. Most patients progress to advanced stages, often with metastasis, resulting in a low survival rate. Despite advancements in surgical and oncological treatments, early diagnosis and better risk stratification remain critical. Methods: This retrospective cross-sectional study focused on analyzing data from patients undergoing pancreaticoduodenectomy for pancreatic adenocarcinoma, in order to determine whether the neutrophil-to-lymphocyte ratio (NLR) and other immune-inflammatory markers, such as the systemic immune-inflammation index (SII) and prognostic nutritional index (PNI), can predict postoperative complications and survival outcomes. Results: Analysis of 136 patients revealed that a higher NLR (≥2.5) was significantly associated with longer overall survival (39 months, IQR: 17-100 months; p = 0.004), compared to lower NLR (<2.5; 18 months, IQR: 9-39 months). Higher SII (≥600) was also associated with significantly improved survival (34 months, IQR: 17-114 months; p = 0.001) compared to lower SII (<600; 20 months, IQR: 9-45 months). No significant differences were observed in postoperative complications or other clinical outcomes between NLR groups, although a trend toward more complications in the higher NLR group was noted (p = 0.06). PNI showed no significant impact on survival (PNI < 38.8: 22 months, IQR: 14-60 months; PNI ≥ 38.8: 33 months, IQR: 14-115 months; p = 0.1) or complications (p = 0.8). Conclusions: Our study highlights the prognostic utility of NLR and SII in patients with adenocarcinoma of the head of the pancreas undergoing pancreaticoduodenectomy. Regarding complications, there were no significant differences across groups stratified by NLR, SII, or PNI, suggesting that while NLR and SII are valuable for predicting long-term oncological outcomes in patients undergoing pancreaticoduodenectomy for adenocarcinoma of the head of the pancreas, they may not be reliable indicators of immediate postoperative morbidity.
Collapse
Affiliation(s)
- Esther Kazlow
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel; (E.K.); (E.R.); (E.G.); (S.M.); (N.E.); (M.N.); (A.A.S.); (W.A.); (R.H.)
- Department of Surgery, Carmel Medical Center, Michal Str. 7, Haifa 3436212, Israel
| | - Elias Rinawi
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel; (E.K.); (E.R.); (E.G.); (S.M.); (N.E.); (M.N.); (A.A.S.); (W.A.); (R.H.)
- Department of Surgery, Carmel Medical Center, Michal Str. 7, Haifa 3436212, Israel
| | - Eden Gerszman
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel; (E.K.); (E.R.); (E.G.); (S.M.); (N.E.); (M.N.); (A.A.S.); (W.A.); (R.H.)
- Department of Surgery, Carmel Medical Center, Michal Str. 7, Haifa 3436212, Israel
| | - Samar Mattar
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel; (E.K.); (E.R.); (E.G.); (S.M.); (N.E.); (M.N.); (A.A.S.); (W.A.); (R.H.)
- Department of Surgery, Carmel Medical Center, Michal Str. 7, Haifa 3436212, Israel
| | - Nabih Essami
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel; (E.K.); (E.R.); (E.G.); (S.M.); (N.E.); (M.N.); (A.A.S.); (W.A.); (R.H.)
- Department of Surgery, Carmel Medical Center, Michal Str. 7, Haifa 3436212, Israel
| | - Mary Nasir
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel; (E.K.); (E.R.); (E.G.); (S.M.); (N.E.); (M.N.); (A.A.S.); (W.A.); (R.H.)
- Department of Surgery, Carmel Medical Center, Michal Str. 7, Haifa 3436212, Israel
| | - Aasem Abu Shtaya
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel; (E.K.); (E.R.); (E.G.); (S.M.); (N.E.); (M.N.); (A.A.S.); (W.A.); (R.H.)
- Department of Gastroenterology, Carmel Medical Center, Haifa 3436212, Israel
| | - Wisam Assaf
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel; (E.K.); (E.R.); (E.G.); (S.M.); (N.E.); (M.N.); (A.A.S.); (W.A.); (R.H.)
- Department of Obstetrics and Gynecology, Carmel Medical Center, Haifa 3436212, Israel
| | - Riad Haddad
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel; (E.K.); (E.R.); (E.G.); (S.M.); (N.E.); (M.N.); (A.A.S.); (W.A.); (R.H.)
- Department of Surgery, Carmel Medical Center, Michal Str. 7, Haifa 3436212, Israel
| | - Ahmad Mahamid
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel; (E.K.); (E.R.); (E.G.); (S.M.); (N.E.); (M.N.); (A.A.S.); (W.A.); (R.H.)
- Department of Surgery, Carmel Medical Center, Michal Str. 7, Haifa 3436212, Israel
| |
Collapse
|
23
|
Dobersch S, Yamamoto N, Schutter A, Cavender SM, Robertson TM, Kartha N, Samraj AN, Doron B, Poole LA, Wladyka CL, Zhang A, Jang GH, Mahalingam AH, Barreto G, Raghavan S, Narla G, Notta F, Eisenman RN, Hsieh AC, Kugel S. HMGA2 and protein leucine methylation drive pancreatic cancer lineage plasticity. Nat Commun 2025; 16:4866. [PMID: 40419509 PMCID: PMC12106622 DOI: 10.1038/s41467-025-60129-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Basal pancreatic ductal adenocarcinoma (PDAC) has the worst overall survival and is the only subtype that serves as an independent poor prognostic factor. We identify elevated levels of LIN28B and its downstream target, HMGA2, in basal PDAC. Notably, LIN28B significantly accelerates KRAS-driven PDAC progression in a mouse model. Here, we show that HMGA2 promotes basal PDAC pathogenesis by enhancing mRNA translation downstream of LIN28B. Mechanistically, HMGA2 suppresses leucine carboxyl methyltransferase 1 (LCMT1) at the chromatin level, reducing PP2A methylation and activity. This leads to increased phosphorylation of S6K and eIF4B, boosting mRNA translation. Additionally, HMGA2 downregulates B56α (PPP2R5A), disrupting functional PP2A holoenzyme assembly and further sustaining phosphorylated S6K levels. Impaired PP2A function mimics HMGA2's effects, reinforcing increased mRNA translation and basal lineage features. This work uncovers a critical link between the LIN28B/HMGA2 axis, protein synthesis, and PDAC lineage specificity via LCMT1-mediated PP2A methylation and B56α-PP2A disruption.
Collapse
Affiliation(s)
| | - Naomi Yamamoto
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
- Molecular & Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA
| | - Aidan Schutter
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Sarah M Cavender
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tess M Robertson
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Nithya Kartha
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Annie N Samraj
- Division of Transfusion Medicine, Department of Laboratory Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Ben Doron
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lisa A Poole
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Cynthia L Wladyka
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Amy Zhang
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Gun Ho Jang
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Guillermo Barreto
- Université de Lorraine, CNRS, Laboratoire IMoPA, Nancy, France
- Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Srivatsan Raghavan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Faiyaz Notta
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Robert N Eisenman
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Andrew C Hsieh
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Sita Kugel
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
| |
Collapse
|
24
|
Wu N, Zhou C, Yan X, Liu Z, Jiang R, Luo Y, Jiang P, Mu Y, Xiao S, Huang X, Zhou Y, Sun D, Jin Y. Integrative Single-Cell and Bulk RNA Sequencing Identifies a Glycolysis-Related Prognostic Signature for Predicting Prognosis in Pancreatic Cancer. Int J Mol Sci 2025; 26:5105. [PMID: 40507914 PMCID: PMC12154285 DOI: 10.3390/ijms26115105] [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: 04/07/2025] [Revised: 05/14/2025] [Accepted: 05/18/2025] [Indexed: 06/16/2025] Open
Abstract
Alterations in glycolysis play a crucial role in cancer cells, influencing tumor aggressiveness and therapeutic effect, particularly in pancreatic adenocarcinoma (PAAD). However, the specific glycolysis-related genes involved in PAAD progression remain poorly understood. This study established glycolysis-related molecular subtypes with distinct survival outcomes using TCGA datasets. The favorable prognosis subtype exhibited enhanced immune infiltration and an activated tumor microenvironment. A glycolysis prognostic model effectively predicted PAAD survival, correlating with global glycolytic pathways, and AUCell evaluated neutrophil communication networks of models. Functional validation demonstrated that ENO1/PGM2L1 co-expression promoted tumor proliferation, migration, invasion, and glycolytic flux in vitro, while accelerating xenograft growth in vivo. Conversely, their knockdown suppressed malignancy. Our study demonstrated that the glycolytic prognostic risk model serves as a reliable tool for prognosis and prediction of PAAD progression. ENO1 and PGM2L1 emerge as key risk factors promoting the malignant progression of PAAD.
Collapse
Affiliation(s)
- Nan Wu
- Laboratory of Medical Genetics, Harbin Medical University, #157 Bao Jian Road, Harbin 150081, China
- Key Laboratory of Preservation of Human Genetics Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Chong Zhou
- Laboratory of Medical Genetics, Harbin Medical University, #157 Bao Jian Road, Harbin 150081, China
- Key Laboratory of Preservation of Human Genetics Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Xu Yan
- Laboratory of Medical Genetics, Harbin Medical University, #157 Bao Jian Road, Harbin 150081, China
- Key Laboratory of Preservation of Human Genetics Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Ziang Liu
- Laboratory of Medical Genetics, Harbin Medical University, #157 Bao Jian Road, Harbin 150081, China
- Key Laboratory of Preservation of Human Genetics Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Ruohan Jiang
- Laboratory of Medical Genetics, Harbin Medical University, #157 Bao Jian Road, Harbin 150081, China
- Key Laboratory of Preservation of Human Genetics Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Yuzhou Luo
- Laboratory of Medical Genetics, Harbin Medical University, #157 Bao Jian Road, Harbin 150081, China
- Key Laboratory of Preservation of Human Genetics Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Ping Jiang
- Laboratory of Medical Genetics, Harbin Medical University, #157 Bao Jian Road, Harbin 150081, China
- Key Laboratory of Preservation of Human Genetics Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Yu Mu
- Laboratory of Medical Genetics, Harbin Medical University, #157 Bao Jian Road, Harbin 150081, China
- Key Laboratory of Preservation of Human Genetics Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Shan Xiao
- Laboratory of Medical Genetics, Harbin Medical University, #157 Bao Jian Road, Harbin 150081, China
- Key Laboratory of Preservation of Human Genetics Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Xien Huang
- Laboratory of Medical Genetics, Harbin Medical University, #157 Bao Jian Road, Harbin 150081, China
- Key Laboratory of Preservation of Human Genetics Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Yunzhen Zhou
- Laboratory of Medical Genetics, Harbin Medical University, #157 Bao Jian Road, Harbin 150081, China
- Key Laboratory of Preservation of Human Genetics Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Donglin Sun
- Laboratory of Medical Genetics, Harbin Medical University, #157 Bao Jian Road, Harbin 150081, China
- Key Laboratory of Preservation of Human Genetics Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
| | - Yan Jin
- Laboratory of Medical Genetics, Harbin Medical University, #157 Bao Jian Road, Harbin 150081, China
- Key Laboratory of Preservation of Human Genetics Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150081, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
25
|
Sherchan A, Jin F, Sherchan B, Mandal SK, Upadhaya Regmi B, Ghising R, Upadhaya SR, Gautam B, Pathak D, Li M. Clinical significance of risk factor analysis in pancreatic cancer by using supervised model of machine learning. Front Med (Lausanne) 2025; 12:1551926. [PMID: 40491770 PMCID: PMC12146380 DOI: 10.3389/fmed.2025.1551926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 04/22/2025] [Indexed: 06/11/2025] Open
Abstract
Introduction Pancreatic cancer (PC) poses a significant global health challenge due to its aggressive nature, late-stage diagnosis, and high mortality despite advancements in treatment. Early detection remains crucial for timely intervention. This study aimed to identify clinically relevant predictors of pancreatic cancer using a supervised machine learning approach and to develop a risk stratification tool with diagnostic capabilities. Methods A matched case-control study was conducted retrospectively at the Tenth People's Hospital of Tongji University (2017-2023), involving 353 cases and 370 matched controls. Demographic and hematological data were extracted from medical records. Variables were pre-selected using cluster dendrograms and subsequently refined using logistic regression with backward elimination and Support Vector Machine (SVM) models. A final risk scoring model was developed based on the best-performing model and internally validated. Results Key predictors retained in the final logistic regression model included Hemoglobin A1c (HbA1c) (OR 1.28; 95% CI: 1.08-1.52), Alkaline Phosphatase (ALP) (OR 1.02; 95% CI: 1.01-1.03), CA19-9 (OR 1.01; 95% CI: 1.01-1.01), Carcinoembryonic Antigen (CEA) (OR 1.41; 95% CI: 1.20-1.66), and Body Mass Index (BMI) (OR 0.88; 95% CI: 0.81-0.97). The final model demonstrated excellent diagnostic performance (AUC = 0.969, p < 0.001), with high accuracy, sensitivity, and specificity. A nomogram was constructed to facilitate individualized PC risk assessment. Conclusion HbA1c, ALP, CA19-9, CEA, and BMI were independently associated with pancreatic cancer. The machine learning-derived risk scoring model demonstrated high predictive accuracy and may serve as a valuable clinical tool for early detection and screening of pancreatic cancer.
Collapse
Affiliation(s)
- Amir Sherchan
- Department of Interventional and Vascular Surgery, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Feng Jin
- Department of Interventional and Vascular Surgery, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bhakti Sherchan
- Department of General Surgery, Scheer Memorial Adventist Hospital, Kavre, Nepal
| | | | | | - Ranita Ghising
- Department of General Surgery, Scheer Memorial Adventist Hospital, Kavre, Nepal
| | | | - Bishnu Gautam
- Department of Radiology, Buddha International Hospital, Dang, Nepal
| | - Dipendra Pathak
- Department of Radiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Maoquan Li
- Department of Interventional and Vascular Surgery, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
26
|
Li D, Rudloff U. Emerging therapeutics targeting tumor-associated macrophages for the treatment of solid organ cancers. Expert Opin Emerg Drugs 2025:1-39. [PMID: 40353504 DOI: 10.1080/14728214.2025.2504376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Over the last decade, immune checkpoint inhibitors (ICIs) like PD-1/PD-L1 or CTLA-4, which reinvigorate T cells for tumor control have become standard-of-care treatment options. In response to the increasingly recognized mechanisms of resistance to T cell activation in immunologically cold tumors, immuno-oncology drug development has started to shift beyond T cell approaches. These include tumor-associated macrophages (TAMs), a major pro-tumor immune cell population in the tumor microenvironment known to silence immune responses. AREAS COVERED Here we outline anti-TAM therapies in current development, either as monotherapy or in combination with other treatment modalities. We describe emerging drugs targeting TAMs under investigation in phase II and III testing with a focus on their distinguishing mechanism of action which include (1) reprogramming of TAMs toward anti-tumor function and immune surveillance, (2) blockade of recruitment, and (3) reduction and ablation of TAMs. EXPERT OPINION Several new immuno-oncology agents are under investigation to harness anti-tumor functions of TAMs. While robust anti-tumor efficacy of anti-TAM therapies across advanced solid organ cancers remains elusive to-date, TAM reprogramming therapies have yielded benefits in select cancers. The inherent heterogeneity of the diverse TAM population will require enhanced investments into biomarker-driven approaches to fully leverage its therapeutic potential.
Collapse
Affiliation(s)
- Dandan Li
- Developmental Therapeutics Branch (TDB), Biology Group, Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Udo Rudloff
- Rare Tumor Initiative, Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
27
|
Lei Y, Liao F, Li X, Zhu Y. Quality and reliability evaluation of pancreatic cancer-related video content on social short video platforms: a cross-sectional study. BMC Public Health 2025; 25:1919. [PMID: 40413453 DOI: 10.1186/s12889-025-23130-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 05/09/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PC) has become one of the leading causes of cancer-related deaths worldwide. Social media platforms are widely used for health information dissemination because of their visual appeal and entertainment value. This study evaluates the content, quality, and reliability of PC-related information on domestic short video platforms. METHODS A total of 265 PC-related videos were retrieved from three short video-sharing platforms: TikTok, Bilibili, and Kwai. The Global Quality Scale (GQS) and the modified DISCERN score were employed to evaluate the quality and content of the videos, respectively. Correlation analysis was conducted to explore the relationships between different video variables. RESULTS The overall quality of the video content was low, with median scores of 2 (IQR: 2-3) for both the GQS and the modified DISCERN score. Most of the videos related to PC were posted by healthcare professionals (219/265, 82.6%). Videos from specialists received more likes on short social video platforms than those from nonspecialists did (median: 678 vs. 270, P = 0.005). Educational videos scored highest in both the GQS (median: 3, IQR 2-3) and the modified DISCERN score (median: 2.5, IQR 2-3). There was a positive correlation between GQS and video duration (r = 0.31, P < 0.01) and the modified DISCERN score (r = 0.434, P < 0.001). CONCLUSION The quality and reliability of the videos on these platforms were generally unsatisfactory in terms of source and content. Videos produced by healthcare professionals or institutions are more informative in terms of comprehensiveness, quality of information, and reliability than are those produced by non-healthcare professionals.
Collapse
Affiliation(s)
- Yuting Lei
- Department of Gastroenterology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yong Waizheng Street, Donghu District, Nanchang, Jiangxi Province, 330006, China
| | - Foqiang Liao
- Department of Gastroenterology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yong Waizheng Street, Donghu District, Nanchang, Jiangxi Province, 330006, China
| | - Xin Li
- Department of Gastroenterology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yong Waizheng Street, Donghu District, Nanchang, Jiangxi Province, 330006, China
| | - Yin Zhu
- Department of Gastroenterology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yong Waizheng Street, Donghu District, Nanchang, Jiangxi Province, 330006, China.
| |
Collapse
|
28
|
Xia Z, Han W, Niu H, Dong H. Global Burden of Pancreatic Cancer Among Individuals Aged 15-59 Years in 204 Countries and Territories, 1990-2021: A Systematic Analysis for the GBD 2021 and Projections to 2045. Cancers (Basel) 2025; 17:1757. [PMID: 40507239 PMCID: PMC12153570 DOI: 10.3390/cancers17111757] [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: 04/03/2025] [Revised: 05/09/2025] [Accepted: 05/16/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PC), the third leading cause of cancer-related mortality globally, exhibits a persistently low five-year survival rate (13%). While the global burden of PC among individuals aged 15-59 years has declined, trends in China remain understudied. This study evaluates global and national trends in PC incidence, mortality, and disability-adjusted life years (DALYs) from 1990 to 2021 and projects trajectories to 2045. METHODS Using data from the Global Burden of Disease (GBD) 2021 study, we calculated age-standardized rates (ASRs) for 204 countries/territories. Joinpoint (version: 5.3.0.0) regression identified temporal trends via average annual percentage changes (AAPCs), and Bayesian age-period-cohort (BAPC) modeling forecasted future burdens. RESULTS Globally, PC burden declined among 15-59-year-olds (AAPC for incidence: -0.8%, 95% UI: -1.2 to -0.4). However, China experienced a significant reversal after 2009, with incidence rising by 1.5% annually (95% UI: 0.9-2.1), disproportionately affecting males. Smoking (contributing to 22.2% of DALYs in China) and high fasting plasma glucose (15%) emerged as key modifiable risk factors, while elevated BMI exacerbated burdens in high SDI regions (3.1% of DALYs). Projections indicate a continued surge in China's PC burden by 2045, particularly among males (incidence projected to increase by 50% from 2010 to 2045). CONCLUSIONS High SDI regions exhibit concentrated PC burdens linked to lifestyle factors, whereas China's rising trends align with healthcare expansion and metabolic disease proliferation. Targeted interventions-smoking cessation, glycemic control, and weight management-are imperative to mitigate growing burdens in younger populations. This study highlights the urgent need for region-specific strategies to address evolving epidemiological challenges in PC prevention and control.
Collapse
Affiliation(s)
- Zeyu Xia
- People’s Hospital of Yiyang, Yiyang 413001, China
| | - Wenping Han
- Teaching and Research Section of Surgery, Faculty of Clinical Medicine, Fenyang College, Shanxi Medical University, Fenyang 032200, China
| | - Haigang Niu
- Teaching and Research Section of Surgery, Faculty of Clinical Medicine, Fenyang College, Shanxi Medical University, Fenyang 032200, China
- Fenyang Hospital of Shanxi Province, Fenyang 032200, China
| | - Hui Dong
- Teaching and Research Section of Surgery, Faculty of Clinical Medicine, Fenyang College, Shanxi Medical University, Fenyang 032200, China
| |
Collapse
|
29
|
Zheng Z, Lu Z, Yan F, Song Y. The role of novel biomarkers in the early diagnosis of pancreatic cancer: A systematic review and meta-analysis. PLoS One 2025; 20:e0322720. [PMID: 40408437 PMCID: PMC12101772 DOI: 10.1371/journal.pone.0322720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/26/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Early detection of pancreatic cancer is essential for improving survival rates. However, noninvasive diagnostic methods are lacking. Novel biomarkers, detectable through liquid biopsy, such as circulating tumor DNA (ctDNA), microRNAs (miRNAs), protein markers, and metabolites, hold promise for early diagnosis. METHODS A systematic search of PubMed, Embase, Web of Science, and the Cochrane Library was conducted for studies published from January 2014 to May 2024. Studies were included if they evaluated novel biomarkers for early pancreatic cancer detection, reported diagnostic performance metrics (sensitivity, specificity), and had a QUADAS-2 score of ≥3. Data on study characteristics, patient demographics, biomarker types, and diagnostic performance were extracted following PRISMA guidelines. A bivariate random-effects model was used to calculate pooled sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), and diagnostic odds ratio (DOR). The area under the summary receiver operating characteristic (SROC) curve assessed overall diagnostic accuracy. The primary outcome was the diagnostic accuracy (sensitivity and specificity) of novel biomarkers in detecting early-stage pancreatic cancer. RESULTS A total of 43 studies involving 19,326 participants were included, with 2,749 patients having stage I or II pancreatic cancer. The pooled sensitivities and specificities were as follows:. miRNA Biomarkers: Sensitivity 0.88 (95% CI 0.79-0.93), Specificity 0.91 (95% CI 0.82-0.95), DOR 72.68 (95% CI 26.64-198.24), AUC 0.95. Protein Biomarkers: Sensitivity 0.79 (95% CI 0.70-0.86), Specificity 0.88 (95% CI 0.82-0.93), DOR 27.74 (95% CI 14.32-53.76), AUC 0.90. Metabolite Biomarkers: Sensitivity 0.84 (95% CI 0.73-0.92), Specificity 0.85 (95% CI 0.81-0.88), DOR 31.76 (95% CI 12.38-81.48), AUC 0.90. ctDNA Biomarkers: Sensitivity 0.65 (95% CI 0.48-0.81), Specificity 0.94 (95% CI 0.88-0.97), DOR 27.73 (95% CI 12.91-59.55), AUC 0.92. Subgroup analyses showed combining biomarkers with CA19-9 improved diagnostic accuracy. Sensitivity analyses confirmed the robustness of the findings. CONCLUSIONS Novel biomarkers, particularly miRNAs and protein markers, demonstrate high diagnostic accuracy for early pancreatic cancer detection and have potential for clinical application in improving early diagnosis and patient outcomes. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero/, Identifier: PROSPERO (CRD42024553633).
Collapse
Affiliation(s)
- Zeyi Zheng
- School of Traditional Chinese Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Ziyu Lu
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Fei Yan
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yani Song
- School of Water Resources and Hydropower Engineering, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
30
|
Jimenez NR, Herman CR, Łaniewski P, Cope E, Lee K, Mahnert ND, Chase DM, Caporaso JG, Herbst-Kralovetz MM. Navigating complexities of polymorphic microbiomes in endometrial cancer. NPJ Biofilms Microbiomes 2025; 11:85. [PMID: 40404643 PMCID: PMC12098703 DOI: 10.1038/s41522-025-00690-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 04/01/2025] [Indexed: 05/24/2025] Open
Abstract
The microbiome is key to understanding endometrial cancer (EC) etiology and prevention strategies, implicated in the regulation of estrogen in estrogen-driven cancers. Utilizing robust methodologies in the QIIME 2 platform, we examined 16S rRNA vaginal and rectal microbiome data from an EC cohort: 192 women with benign gynecologic conditions, endometrial hyperplasia, or endometrial cancer. Distinct microbial compositions and community networks specific to EC were identified and related to histological grade with adjustments for EC risk factors. Vaginal health-associated Lactobacillus and Limosilactobacillus, and rectal Prevotella and Peptoniphilus, were depleted in EC, while detrimental vaginal Anaerococcus, Porphyromonas, Prevotella, Peptoniphilus, and rectal Buttiaxella were enriched. Significant bacterial features were shared between rectal and vaginal sites in EC, such as Prevotella timonensis and Peptoniphilus A. Vaginal Lactobacillus abundance contributed to less feature sharing from the rectum. Putative microbial metabolic analysis identified dysregulation of amino acid, complex carbohydrate, and hormone metabolism amongst patients with EC.
Collapse
Affiliation(s)
- Nicole R Jimenez
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Chloe R Herman
- Center for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
- School of Informatics, Computing and Cyber Systems, Northern Arizona University, Flagstaff, AZ, USA
| | - Paweł Łaniewski
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Emily Cope
- Center for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Keehoon Lee
- Translational Genomics Research Institute, part of City of Hope, Flagstaff, AZ, USA
| | - Nichole D Mahnert
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Dana M Chase
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - J Gregory Caporaso
- Center for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA.
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
| | - Melissa M Herbst-Kralovetz
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
| |
Collapse
|
31
|
Chen Y, Zhang F, Dai S, Zhao J, Cai W, Zhang K, Liao X, Chen L. Lactate-associated gene MCU promotes the proliferation, migration, and invasion of pancreatic ductal adenocarcinoma. BMC Cancer 2025; 25:913. [PMID: 40399795 PMCID: PMC12096505 DOI: 10.1186/s12885-025-14319-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND The metabolism of lactate and lactylation of proteins are believed to influence tumor development through their effects on the tumor microenvironment and immune escape mechanisms. Nevertheless, its significance in pancreatic ductal adenocarcinoma (PDAC) has yet to be fully understood. This investigation sought to assess the predictive value and treatment implications of lactate-related genes (LRGs) in PDAC. METHODS We analyzed PDAC data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), identifying LRGs. Using weighted gene co-expression network analysis (WGCNA) and consensus clustering, we delineated lactate subtypes and extracted differentially expressed genes. Functional enrichment and gene set enrichment analysis (GSEA) analyses were conducted to explore pathways. A lactate-linked risk signature was constructed using Lasso-Cox regression, and its prognostic value was validated. In vitro experiments were executed to examine the function of MCU in PDAC cells. In vitro experiments were conducted to detect the malignant potential of MCU in PDAC cells and its effect on lactic acid metabolism. RESULTS Two lactate subtypes were identified, with distinct gene expression profiles and clinical outcomes. The risk signature, comprising four LRGs, predicted survival with significant accuracy. In vitro, MCU knockdown reduced cell proliferation, migration, invasion, and stemness, confirming its role in PDAC malignancy. At the same time, it can also inhibit lactate production and glycolysis processes. CONCLUSION Our investigation underscores the importance of LRGs in PDAC, providing a novel prognostic signature and therapeutic target.
Collapse
Affiliation(s)
- Yuhang Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Fenglin Zhang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Graduate School of Anhui, University of Traditional Chinese Medicine, Hefei, 230022, China
| | - Suoyi Dai
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jiangang Zhao
- Department of Oncology, Shaoxing Central Hospital, Shaoxing, 312030, China
- Department of Oncology, The Central Affiliated Hospital, Shaoxing University, Shaoxing, 312030, China
| | - Wenxun Cai
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ke Zhang
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China
| | - Xinghe Liao
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Lianyu Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
32
|
Hao Z, Zhang H, Dang Y, Qiu J, Yan M, Yan X, Huang Z, Ren C, Zhang T, Wu W, Huo L. Feasibility of simplified Patlak parametric imaging with scaled population-based input function on pancreatic cancer. EJNMMI Phys 2025; 12:46. [PMID: 40392375 PMCID: PMC12092890 DOI: 10.1186/s40658-025-00758-z] [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: 09/17/2024] [Accepted: 04/30/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND This study evaluates the feasibility of using a simplified Patlak parametric imaging technique with a scaled population-based input function (sPBIF) in pancreatic cancer. METHODS Twenty-six patients underwent multi-bed, multi-pass [18F]FDG PET/CT scans, from which both dynamic and static PET images were reconstructed. Patlak parametric images were generated from the dynamic PET series using both the image-derived input function (IDIF) and the sPBIF. The consistency between IDIF and sPBIF was evaluated by comparing the area under the curve (AUC) and Patlak parameters derived from both input functions. The detectability of pancreatic lesions, assessed by tumor-to-background ratio (TBR) and contrast-to-noise ratio (CNR), was compared between SUV and Patlak parametric images. Additionally, the correlation between clinicopathological features and PET parameters, including SUV and Patlak values, was analyzed. RESULTS We found good agreement between the AUC for IDIF and sPBIF with correlation coefficients of 0.87 and 0.93 for the 0-30 min and 0-50 min intervals, respectively. The Patlak parameters from IDIF and sPBIF presented correlation coefficients higher than 0.94. The SUV and Patlak Ki exhibited correlation coefficients greater than 0.92 and 0.73 in malignant and benign pancreatic lesions, respectively. The SUV and Patlak V0 correlated with correlation coefficients higher than 0.75 in benign lesions, but exhibited only a weak correlation in malignant lesions. The TBR of Patlak Ki was significantly higher in malignant lesions compared to SUV. However, the CNR of Patlak Ki was lower due to increased noise in the parametric images. Most clinicopathological features showed weak correlation with PET parameters, except for a marginal classification of lesion differentiation by the maximum Ki value. CONCLUSIONS The sPBIF approach enables the acquisition of additional Patlak parametric images alongside static SUV imaging in pancreatic cancer patients. Ki parametric imaging provided higher contrast than static imaging for detecting pancreatic lesions.
Collapse
Affiliation(s)
- Zhixin Hao
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Haiqiong Zhang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Yonghong Dang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Mengshi Yan
- Beijing United Imaging Research Institute of Intelligent Imaging, Beijing, 100094, China
| | - Xinchun Yan
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Zhenghai Huang
- Medical Science Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Chao Ren
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China.
| | - Wenming Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China.
| | - Li Huo
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Center for Rare Diseases Research, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
33
|
Xu L, Wan X, Shan X, Zha W, Shi Y, Fan R. ONECUT3 activates the TRIM46-NF-κB pathway to promote the development of pancreatic cancer. Biochem Biophys Res Commun 2025; 759:151705. [PMID: 40154001 DOI: 10.1016/j.bbrc.2025.151705] [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: 11/27/2024] [Revised: 02/24/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Pancreatic cancer (PC) remains one of the deadliest cancers, characterized by its high aggressiveness and low overall survival, with chemotherapy and immunotherapy showing limited efficacy. It is essential to investigate the molecular mechanisms driving the PC progression. In this study, we showed that One Cut homeobox 3 (ONECUT3) acted as an oncogene promoting PC progression and observed a significant increase of ONECUT3 levels in PC tissues and cells. The reduced ONECUT3 expression was positively correlated with decreased tumor volumes and weight, and the depressed proliferation, migration and invasion abilities. Mechanistically, ONECUT3 directly bound to the promoter of tripartite motif-containing 46 (TRIM46) and transcriptionally upregulated its expression. Tripartite motif (TRIM)-containing proteins have been identified as closely linked to the advancement of tumors. However, the role of TRIM46 in PC remains largely unexplored. The expression of ONECUT3 was found to be positively linked with TRIM46 in human PC tissues. The upregulation of TRIM46 rescued ONECUT3 knockdown-induced suppression of cell proliferation, migration and invasion abilities, and tumor growth in PC. TRIM46 overexpression also activated NF-κB signaling in PC cells. To sum up, ONECUT3 has been identified as a promising prognostic indicator in PC, and targeting this cancer-promoting pathway could offer an effective therapeutic approach to combat the PC progression.
Collapse
Affiliation(s)
- Linyi Xu
- The Yancheng Clinical College of Xuzhou Medical University, 224001, China
| | - Xinqiang Wan
- Department of Gynaecology and Obstetrics, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, No.166, Yulong West Road, Yancheng, Jiangsu Province, 224001, China
| | - Xiangxiang Shan
- Department of Geraeology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, No.166, Yulong West Road, Yancheng, Jiangsu Province, 224001, China
| | - Wenzhang Zha
- Department of General Surgery, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, No.166, Yulong West Road, Yancheng, Jiangsu Province, 224001, China
| | - Yuhua Shi
- Department of General Surgery, Affiliated Hospital of Nantong University, Third People's Hospital of Yancheng, No.75, Juchang Road, Yancheng, Jiangsu Province 224001, China.
| | - Rengen Fan
- Department of General Surgery, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, No.166, Yulong West Road, Yancheng, Jiangsu Province, 224001, China.
| |
Collapse
|
34
|
Ducreux M, Desgrippes R, Rinaldi Y, Di Fiore F, Guimbaud R, Evesque L, Bachet JB, Vanelslander P, Lecomte T, Capitain O, Parzy A, Bolliet M, Etienne PL, Forestier J, El Hajbi F, Bignon AL, Lebrun-Ly V, De Sousa Carvalho N, Texier M, Bouche O. PRODIGE 29-UCGI 26 (NEOPAN): A Phase III Randomized Trial Comparing Chemotherapy With FOLFIRINOX or Gemcitabine in Locally Advanced Pancreatic Carcinoma. J Clin Oncol 2025:JCO2402210. [PMID: 40378359 DOI: 10.1200/jco-24-02210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 03/11/2025] [Accepted: 04/03/2025] [Indexed: 05/18/2025] Open
Abstract
PURPOSE More than 30% of patients with pancreatic cancer are unresectable because of the local extension with a median overall survival (OS) of <1 year. Combination of fluorouracil (FU), oxaliplatin, and irinotecan (FOLFIRINOX) is superior to gemcitabine in the treatment of metastatic pancreatic cancer, but standard of care remains gemcitabine in locally advanced pancreatic cancer (LAPC). METHODS Patients with histologically proven LAPC not suitable for surgery, Eastern Cooperative Oncology Group WHO performance status (PS) ≤1 were eligible. Random assignment was stratified by center, tumor localization (pancreas head yes/no), WHO PS (0 v 1), and age (≤60 years v >60 years). Patients received FOLFIRINOX or gemcitabine for 6 months. The primary end point was progression-free survival (PFS). Main secondary end points were OS, time to treatment failure, quality of life, and safety. One hundred seventy patients (142 events) were needed to detect an increase of 3 months in PFS with 80% power (log-rank test, 5% two-sided α). RESULTS One hundred seventy one patients age 35-84 years were included and followed for a maximum of 5 years. With a median follow-up of 59.6 months (95% CI, 42.3 to not reached), 168 events were observed and the median PFS was 9.7 months (95% CI, 7.0 to 11.7) with FOLFIRINOX versus 7.7 months (95% CI, 6.2 to 9.2) with gemcitabine, hazard ratio (HR), 0.7 (95% CI, 0.5 to 1.0), P = .04. The median OS was 15.7 months (95% CI, 11.9 to 20.4) in the FOLFIRINOX group versus 15.4 months (95% CI, 11.7 to 18.6) in the gemcitabine group, HR, 1.02 (95% CI, 0.73 to 1.43), P = .95. CONCLUSION Results confirm that FOLFIRINOX improves PFS significantly compared with gemcitabine and is well tolerated in LAPC. No significant difference in OS was observed between both groups.
Collapse
Affiliation(s)
- Michel Ducreux
- Gustave Roussy Cancer Center, Tumor Cells Dynamics, INSERM U1279, Université Paris-Saclay, Villejuif, France
| | - Romain Desgrippes
- Hepato-Gastroenterology and Digestive Oncology Department, Centre Hospitalier de Saint Malo, Saint Malo, France
| | | | - Frédéric Di Fiore
- Gastroenterology, CHU Hôpitaux de Rouen-Charles Nicolle, Rouen, France
| | - Rosine Guimbaud
- Digestive Oncology, Centre Hospitalier Rangueil, Toulouse, France
| | | | - Jean-Baptiste Bachet
- Sorbonne University, Hepato-Gastroenterology Department, Groupe Hospitalier Pitié Salpetrière, APHP, Paris, France
| | | | - Thierry Lecomte
- Hepatogastroenterology and Digestive Oncology Departement, Hôpital Trousseau, Chambray-Les-Tours, France
| | - Olivier Capitain
- Medical Oncology, Institut de Cancérologie de l'Ouest-Centre Paul Papin, Angers, France
| | - Aurélie Parzy
- Digestive Pathology, Centre Francois Baclesse, Caen, France
| | - Marion Bolliet
- Gastroenterology, Hôpitaux Civils de Colmar, Colmar, France
| | - Pierre-Luc Etienne
- Medical Oncology, Hôpital Privé des Côtes d'Armor, Plerin sur Mer, France
| | | | - Farid El Hajbi
- Urology and Digestive Oncology, Centre Oscar Lambret, Lille, France
| | | | | | | | - Matthieu Texier
- Biostatistics, Gustave Roussy Institut de Cancérologie, Villejuif, France
| | | |
Collapse
|
35
|
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
|
36
|
Earl J. A plasma-based multimetabolite biomarker for early detection of pancreatic cancer. Lancet Gastroenterol Hepatol 2025:S2468-1253(25)00089-5. [PMID: 40388949 DOI: 10.1016/s2468-1253(25)00089-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 05/21/2025]
Affiliation(s)
- Julie Earl
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Ramón y Cajal Health Research Institute (IRYCIS), 28034 Madrid, Spain.
| |
Collapse
|
37
|
Akhtar MA, Chowdhury IR, Taneja B. Healthcare utilisation and economic burden of cancer on Indian households. Sci Rep 2025; 15:16780. [PMID: 40368978 PMCID: PMC12078705 DOI: 10.1038/s41598-025-01279-6] [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: 09/11/2024] [Accepted: 05/05/2025] [Indexed: 05/16/2025] Open
Abstract
India is experiencing a rising incidence of cancer with a high mortality rate at a younger age. With high catastrophic healthcare expenditures and inadequate social security, it is imperative to investigate the health-seeking behaviour and corresponding economic burden on cancer-affected households. Using a nationally representative database, we use the matching methods to analyse healthcare utilization, healthcare expenditure, and financial distress of cancer-affected households. We find that the cancer-affected households report a longer hospital stay, more surgery, medicine uptake, and diagnostic tests per member compared to non-cancer households. The out-of-pocket healthcare expenditure borne by the affected households are significantly higher and mostly inflated through inpatient care. Further, we find some adverse spill-over effect in terms of lower per-member inpatient and outpatient visits and lower healthcare expenditure for non-cancer members of affected households when they are afflicted with any diseases. The catastrophic expenditures inflict income loss and distress financing to the cancer affected households. Further, we find per-member work force participation and non-medical consumption expenditures significantly low. We also observe heterogeneous effect in terms of lower inpatient visits, lower medicine uptake, lower capability to pay for treatment, and significantly higher borrowing and selling off assets among various cancer affected socioeconomically disadvantaged groups.
Collapse
Affiliation(s)
| | - Indrani Roy Chowdhury
- Centre for the Study of Regional Development, Jawaharlal Nehru University, New Delhi, India.
| | - Bhawna Taneja
- Centre for the Study of Regional Development, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
38
|
Demyan L, Weiss MJ. Personalized Care for Pancreatic Cancer: Harnessing Patient-Derived Organoids. J Gastrointest Cancer 2025; 56:113. [PMID: 40347361 DOI: 10.1007/s12029-025-01164-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/01/2025] [Indexed: 05/12/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most fatal cancers. Surgical resection combined with appropriate chemotherapy currently offers the best chance for long-term survival and potential cure. However, effective treatment is hindered by the limited chemotherapy options and the absence of reliable clinical tools to guide chemotherapy selection. Patient-derived organoids (PDOs) have emerged as a promising technology with the potential in precision medicine for PDAC. This review provides an overview of pancreatic organoid genesis, explores the role of PDOs in elucidating PDAC biology within clinically relevant contexts, and concludes by examining current literature on the utility of PDOs as biomarkers for personalized treatment strategies.
Collapse
Affiliation(s)
- L Demyan
- Northwell Health, New Hyde Park, USA.
| | - M J Weiss
- Northwell Health, New Hyde Park, USA.
- Donald & Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, USA.
| |
Collapse
|
39
|
Jia Y, Li J, Mei W, Zhang H, Wang Z, Xie X, Gao C, Xu X, Li F. Pan-HDAC inhibitor LAQ824 inhibits the progression of pancreatic ductal adenocarcinoma and suppresses immune escape by promoting antigen presentation. Int Immunopharmacol 2025; 154:114528. [PMID: 40158429 DOI: 10.1016/j.intimp.2025.114528] [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: 01/26/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025]
Abstract
Pancreatic cancer is the seventh leading cause of cancer-related deaths worldwide, with a dismal 5-year survival rate. New drugs targeting pancreatic ductal adenocarcinoma (PDAC), the primary pathological subtype, are urgently needed. LAQ824, a novel pan-histone deacetylase inhibitor (HDACi), has shown anti-tumor activity in various cancers, but its effects on PDAC remain unexplored. This study investigates the therapeutic potential of LAQ824 in PDAC and its role in modulating immune escape mechanisms. Using a subcutaneous tumor model in C57BL/6 J mice, LAQ824's anti-tumor effects were evaluated. In vitro and in vivo experiments-including IHC, flow cytometry, RNA sequencing, and single-cell RNA sequencing-demonstrated that LAQ824 inhibits tumor proliferation, suppresses the epithelial-mesenchymal transition (EMT), and induces apoptosis. LAQ824 also enhances immunogenicity by upregulating MHC-I-mediated antigen presentation, increasing immune cell infiltration, and promoting CD8+ T cell maturation and differentiation. Mechanistically, LAQ824 upregulated MHC-I expression by enhancing chromatin accessibility of related genes, with HDAC1 identified as a key repressor of MHC-I in PDAC cells. In conclusion, we found that LAQ824 has a significant anti-tumor effect in PDAC. LAQ824 not only directly affects general biological behaviors such as proliferation, apoptosis, and EMT, but also increases the immunogenicity of tumor cells by upregulating the expression of MHC-I in PDAC, which promotes the antigen presentation process and enhances anti-tumor immunity. By showcasing LAQ824's potential as a therapeutic target against PDAC, the present study provides novel insights into the link between epigenetic regulation and immunogenicity in PDAC.
Collapse
Affiliation(s)
- Yuchen Jia
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Beijing, China
| | - Jie Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Beijing, China
| | - Wentong Mei
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Beijing, China; Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Haoyu Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Beijing, China
| | - Zheng Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Beijing, China
| | - Xiaozhou Xie
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Beijing, China
| | - Chongchong Gao
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Beijing, China.
| | - Xiaoqing Xu
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Beijing, China.
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Beijing, China.
| |
Collapse
|
40
|
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
|
41
|
Yonemura H, Kuwatani M, Nakajima K, Masamune A, Ogawa M, Sakamoto N. Treatment of Pancreatic Cancer Using Near-Infrared Photoimmunotherapy Targeting Cancer-Associated Fibroblasts in Combination with Anticancer Chemotherapeutic Drug. Cancers (Basel) 2025; 17:1584. [PMID: 40361512 PMCID: PMC12071749 DOI: 10.3390/cancers17091584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/26/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC), which has a poor prognosis, involves an overabundance of fibroblasts and extracellular matrix. Cancer-associated fibroblasts (CAFs) are critical for providing structural support by secreting soluble factors and extracellular matrix proteins into the stroma. We assessed the potential of near-infrared photoimmunotherapy (NIR-PIT) targeting CAFs in PDAC. Methods: PDAC cells (Capan-1 and SUIT-2) and CAFs (hPSC-5) were used. Anti-human fibroblast activation protein (FAP)/podoplanin (PDPN) antibodies were used to bind to CAFs and conjugates with the specific photosensitizer IRDye®700DX (IR700) to investigate the effects of NIR-PIT. Thereafter, BALB/c Slc-nu/nu mice were transplanted with Capan-1 and/or CAFs and treated with gemcitabine (GEM) with or without NIR-PIT. Results: The binding rate of anti-FAP antibody-AlexaFluor®488 conjugate to hPSC-5 cells was high, whereas that of the anti-PDPN antibody-conjugate was not. The incubation of anti-FAP antibody-IR700 conjugate (αFAP-IR700) with hPSC-5 cells for 3 h led to maximal fluorescence on the surface of hPSC-5 cells. When NIR-PIT with αFAP-IR700 was performed in the co-culture group of Capan-1 and hPSC-5 cells, the proliferative capacity of Capan-1 cells decreased to the same level as that when Capan-1 cells were cultured alone (p < 0.05). In vivo, compared with the GEM group, the NIR-PIT with the GEM group showed a significant reduction in the tumor volume (day 28: 79 vs. 382 mm3, p < 0.05). Tumor volumes in the NIR-PIT group were not reduced compared with those in the control group. Conclusions: Combining NIR-PIT with conventional chemotherapy to target CAFs may enhance the anticancer effects on PDAC.
Collapse
Affiliation(s)
- Hiroki Yonemura
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, North 15, West 7, Kita-ku, Sapporo 060-8648, Hokkaido, Japan; (H.Y.); (N.S.)
| | - Masaki Kuwatani
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, North 15, West 7, Kita-ku, Sapporo 060-8648, Hokkaido, Japan; (H.Y.); (N.S.)
| | - Kohei Nakajima
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, North 12, West 6, Kita-ku, Sapporo 060-0812, Hokkaido, Japan; (K.N.); (M.O.)
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Hospital, Seiryocho1-1, Aoba-ku, Sendai 980-8574, Miyagi, Japan;
| | - Mikako Ogawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, North 12, West 6, Kita-ku, Sapporo 060-0812, Hokkaido, Japan; (K.N.); (M.O.)
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, North 21, West 10, Kita-ku, Sapporo 001-0021, Hokkaido, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, North 15, West 7, Kita-ku, Sapporo 060-8648, Hokkaido, Japan; (H.Y.); (N.S.)
| |
Collapse
|
42
|
Walczak ŁJ, Kosikowska U, Herbet M. The role and significance of the oncobiota in selected cancers: a review. Clin Exp Med 2025; 25:141. [PMID: 40335827 PMCID: PMC12058861 DOI: 10.1007/s10238-025-01598-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: 11/18/2024] [Accepted: 02/10/2025] [Indexed: 05/09/2025]
Abstract
This review provides an overview of research evidence focused on the microbial components essential to clinical cancer care, called the oncobiota (the interaction of human microbiota and cancer cells). It specifically examines the oncobiota in central nervous system cancer,breast cancer, pancreatic cancer, liver cancer, lung cancer, and cervical cancer. The literature review reveals insufficient knowledge about the oncobiota of organs once considered sterile. Many studies on oncobiota focus on small, geographically specific patient groups, and the absence of a reference (control) group complicates the development of microbial profiles for selected cancers. Consequently, this review aims to analyze the literature data and reports on the role of oncobiota in selected "sterile" organs and the resulting therapeutic or preventive implications. All relevant publications on oncobiota in patients with the selected cancers were considered to provide the most thorough analysis possible. Understanding the significance and role of oncobiota in the pathomechanisms of carcinogenesis may pave the way for targeted cancer prevention methods. Furthermore, therapeutic strategies based on oncobiota could represent a novel area of personalized cancer treatment. Additionally, oncobiota may serve as an additional diagnostic tool in oncology.
Collapse
Affiliation(s)
- Łucja Justyna Walczak
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 8 Chodźki Street, 20-093, Lublin, Poland.
| | - Urszula Kosikowska
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Medical University of Lublin, 1 Chodźki Street, 20-093, Lublin, Poland.
| | - Mariola Herbet
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 8 Chodźki Street, 20-093, Lublin, Poland
| |
Collapse
|
43
|
Ye J, Qin SS, Hughson AL, Hannon G, Salama NA, Vrooman TG, Lesch ML, Lesser S, Eckl SL, Jewell R, Benoodt L, Mills BN, Johnston CJ, Lord E, Belt BA, Calvi LM, Linehan D, Luheshi N, Eyles J, Gerber SA. Blocking LIF and PD-L1 enhances the antitumor efficacy of SBRT in murine PDAC models. J Immunother Cancer 2025; 13:e010820. [PMID: 40341024 PMCID: PMC12067785 DOI: 10.1136/jitc-2024-010820] [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: 10/13/2024] [Accepted: 04/21/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Recent preclinical and clinical data suggest that leukemia inhibitory factor (LIF) is a potential target for various tumor types including pancreatic ductal adenocarcinoma as LIF is involved in multiple protumor processes including cancer stem cell maintenance, epithelial-mesenchymal transition (EMT), immunosuppression, and chemo/radioresistance. Anti-LIF antibody therapy has demonstrated safety and tolerability but limited efficacy in phase 1 clinical trial in advanced solid tumors. This prompted us to explore combination therapies, suggesting that LIF blockade, when combined with standard-of-care chemotherapy, radiotherapy, and/or immunotherapy, could present a promising therapeutic strategy. METHODS We evaluated the impact of combining systemic inhibition of LIF/programmed death-ligand 1 (PD-L1) with localized stereotactic body radiotherapy (SBRT) on tumor progression across multiple murine orthotopic pancreatic tumor models and examined systemic antitumor immunity using a hepatic rechallenge model. The antitumor immune response was characterized throughflow cytometry and Luminex assays. To identify differentially expressed genes and signaling pathways following treatment, we performed bulk RNA sequencing on pancreatic tumors. Additionally, single-cell RNA sequencing was conducted to further examine changes in tumor-infiltrating immune cells and their signaling pathways. RESULTS We showed that simultaneous inhibition of LIF and PD-L1 significantly amplified the antitumor efficacy of SBRT, resulting in extended survival. The triple therapy (SBRT+anti-LIF+anti-PD-L1) generated an immunostimulatory tumor microenvironment, characterized by a proinflammatory shift in the cytokine/chemokine profile, increased infiltration of effector CD8+ T cells, and upregulated activation or maturation signals in tumor-infiltrating CD8+ T cells and macrophages. The beneficial effects of triple therapy were mostly abrogated by depletion of CD8+ T cells. In addition, triple therapy downregulated pathways related to tumor stemness, proliferation, and metabolism, and reduced EMT. Importantly, the combination of local SBRT treatment with systemic LIF and PD-L1 blockade resulted in long-term systemic antitumor memory.
Collapse
Affiliation(s)
- Jian Ye
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Shuyang S Qin
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Angela L Hughson
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Gary Hannon
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Noah A Salama
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Tara G Vrooman
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Maggie L Lesch
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Sidney Lesser
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Sarah L Eckl
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Rachel Jewell
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Lauren Benoodt
- Genomic Research Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Bradley N Mills
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Carl J Johnston
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Edith Lord
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Brian A Belt
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Laura M Calvi
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
- Department of Medicine Hematology/Oncology, University of Rochester Medical Center, Rochester, New York, USA
| | - David Linehan
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Nadia Luheshi
- The Discovery Center, Cambridge Biomedical Campus, Cambridge, UK
| | - Jim Eyles
- Oncology R&D, Research and Early Development, AstraZeneca R&D, Cambridge, Cambridgeshire, UK
| | - Scott A Gerber
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
44
|
Pei K, Li D, Xi M. The impact of lymphadenectomy on cancerspecific survival in patients with low-grade endometrioid carcinoma of stage T1a. Sci Rep 2025; 15:15952. [PMID: 40335549 PMCID: PMC12059127 DOI: 10.1038/s41598-025-00531-3] [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: 02/25/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
For patients with low-grade endometrioid carcinoma of stage T1a, the role of lymphadenectomy in staging surgery remains controversial. This study aims to evaluate the impact of lymphadenectomy on cancer-specific survival (CSS) in this patient population using a large, population-based dataset. We conducted a retrospective analysis using the Surveillance, Epidemiology, and End Results (SEER) database, identifying 11,014 patients with stage T1a, low-grade endometrioid carcinoma from 2004 to 2015. Patients were divided into lymphadenectomy and non-lymphadenectomy groups. Propensity score matching (PSM) was performed to balance baseline characteristics. Kaplan-Meier analysis, log-rank tests, and multivariate Cox regression were used to assess CSS and identify independent prognostic factors. Before PSM, the non-lymphadenectomy group had higher CSS compared to the lymphadenectomy group (HR = 1.56, 95% CI: 1.16-2.10, p = .003). After 1:1 PSM, CSS was similar between the two groups (HR = 1.09, 95% CI: 0.78-1.53, p = .605). Subgroup analyses showed no significant differences in CSS except for the subgroup with tumor size > 2 cm, where non-lymphadenectomy was associated with better CSS (HR = 0.50, p = .035). Multivariate Cox regression analysis identified age, marital status, histological grade, and chemotherapy as independent prognostic factors for CSS, while lymphadenectomy was not (p = .980).. Our findings suggest that lymphadenectomy does not improve CSS in patients with low-grade endometrioid carcinoma of stage T1a.
Collapse
Affiliation(s)
- Kaige Pei
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, No. 20, Section 3, Renmin South Road, Wuhou District, Chengdu, 610041, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Dongmei Li
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, No. 20, Section 3, Renmin South Road, Wuhou District, Chengdu, 610041, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Mingrong Xi
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, No. 20, Section 3, Renmin South Road, Wuhou District, Chengdu, 610041, Sichuan, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China.
| |
Collapse
|
45
|
Yu H, Zu L, Zang Y, Teng F, Wang T, Wu M, Wang Y, Xue F. Assessment of NUDT5 in Endometrial Carcinoma: Functional Insights, Prognostic and Therapeutic Implications. Biomedicines 2025; 13:1136. [PMID: 40426963 PMCID: PMC12108576 DOI: 10.3390/biomedicines13051136] [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: 02/16/2025] [Revised: 03/09/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Endometrial carcinoma (EC) is the most common gynecological malignancy, with increasing incidence contributing to a significant global health burden. Despite recent advancements, the molecular mechanisms underlying EC progression remain insufficiently understood, limiting the development of targeted therapies. This study aims to investigate the role of nucleoside diphosphate-linked moiety X motif 5 (NUDT5) in EC and evaluate its potential as a biomarker and therapeutic target. Methods: This study analyzed gene expression data from The Cancer Genome Atlas and performed tissue microarray validation to assess NUDT5 expression in EC samples. Immunohistochemistry was used to evaluate NUDT5 protein levels and their correlation with clinicopathological features. Functional assays, including cell proliferation, migration, invasion, and apoptosis analysis, were conducted to determine the oncogenic effects of NUDT5 in vitro. Weighted gene co-expression network analysis (WGCNA) and experimental validation were performed to explore the impact of NUDT5 on the PI3K-AKT signaling pathway, while tumor growth assays in xenograft models assessed the therapeutic potential of NUDT5 inhibition in vivo. Results: NUDT5 was significantly overexpressed in EC tissues and correlated with advanced histological grade and poor prognosis. Functional experiments demonstrated that NUDT5 promotes cell proliferation, migration, and invasion while inhibiting apoptosis. Mechanistically, NUDT5 activated the PI3K-AKT pathway, contributing to tumor progression. In vivo, NUDT5 knockdown suppressed tumor growth. Conclusions: These findings suggest that NUDT5 functions as an oncogene in EC, serving as a potential diagnostic and prognostic biomarker. Targeting NUDT5 may provide a novel therapeutic strategy for EC management.
Collapse
Affiliation(s)
- Hongfei Yu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
| | - Lingling Zu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
| | - Yuqin Zang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
| | - Fei Teng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
| | - Tao Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
| | - Ming Wu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300070, China
| |
Collapse
|
46
|
Earl J, Kataki A, Canzian F, Costello E, Campa D, Greenhalf W. Targeting pancreatic cancer screening by identification of pathogenic variants of BRCA2/ BRCA1 in healthy individuals who have no known family history of pancreatic cancer: The arguments for and against. Semin Cancer Biol 2025; 113:1-8. [PMID: 40339999 DOI: 10.1016/j.semcancer.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/15/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
The majority of patients with pancreatic ductal adenocarcinoma (PDAC) are no longer suitable for treatment with curable intent at the time of diagnosis resulting in a 5-year survival of less than 10 %. Imaging of asymptomatic individuals could identify early cancers, but only with a risk of falsely identifying a benign lesion as malignant. Screening of an unselected population would result in far more such false positives than true early cancers. Selection before screening is therefore essential, but there are very few populations at high enough risk to make screening more beneficial than counterproductive. These populations include carriers of specific mutations in BRCA2, and arguably BRCA1, who have a family history of PDAC. These pathogenic mutations all have a predictable effect in making loss of Homologous Recombination Repair (HRR) likely in a carrier's lifetime. In this review the impact of such loss of HRR function on the likelihood of PDAC development will be discussed. Furthermore, it will be discussed whether the identification of a germline pathogenic mutation is sufficient to justify carrier surveillance for the development of the malignancy, or whether the current practice of screening only those carriers with a close relative diagnosed with PDAC is justifiable, as only a proportion of carriers are at high risk. The review will go beyond this to discuss whether there is an essential need to better define and stratify those at high risk, so that only high-risk carriers are put on surveillance.
Collapse
Affiliation(s)
- Julie Earl
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, Carretera Colmenar Km 9,100, Madrid 28034, Spain
| | - Agapi Kataki
- Department of Propaeudeutic Surgery, Breast Unit, Medical School, National and Kapodistrian University of Athens, Greece
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | | | - William Greenhalf
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK.
| |
Collapse
|
47
|
Zhang Y, Esmail A, Hassanain H, Dhillon V, Abdelrahim W, Al-Najjar E, Khasawneh B, Abdelrahim M. Correlative Analysis of Tumor-Informed Circulating Tumor DNA (ctDNA) and the Survival Outcomes of Patients with Pancreatic Adenocarcinoma. Biomedicines 2025; 13:1124. [PMID: 40426952 PMCID: PMC12108567 DOI: 10.3390/biomedicines13051124] [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: 03/28/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with poor prognosis due to late-stage diagnosis, limited surgical resectability, and frequent recurrence. Traditional biomarkers like CA19-9 and imaging techniques often fail to detect minimal residual disease (MRD) or early recurrence. Circulating tumor DNA (ctDNA) is a promising non-invasive biomarker that may provide early detection of disease recurrence, offering a potential improvement in patient management. This study aimed to assess the utility of ctDNA as a prognostic tool for PDAC patients, specifically in predicting recurrence and overall survival (OS). Methods: This retrospective study analyzed data from 39 PDAC patients who underwent surgery and were monitored for ctDNA levels using Signatera™, a tumor-informed multiplex PCR next-generation sequencing assay. Blood samples were collected both preoperatively and postoperatively, and ctDNA levels were measured to detect MRD. The sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) of ctDNA were compared with CA19-9 in detecting disease recurrence. Clinical outcomes, including progression-free survival (PFS) and OS, were evaluated in relation to ctDNA status. Results: Among 39 patients, 153 plasma samples were analyzed, with 17 patients testing positive for ctDNA. Sensitivity of ctDNA in detecting relapse was 91%, compared to 83% for CA19-9, with combined testing reaching 98% sensitivity. ctDNA positivity was associated with significantly shorter OS and PFS, with patients testing negative for ctDNA having a median OS of 37.6 months versus 13.4 months in ctDNA-positive patients (p = 0.003). The median time from ctDNA positivity to imaging-confirmed relapse was 81 days. Positive ctDNA was also linked to higher rates of lymphovascular invasion and positive surgical margins, highlighting the aggressive nature of the disease in these patients. Conclusions: CtDNA is a highly sensitive and specific biomarker for detecting MRD and predicting recurrence in PDAC patients, offering superior performance over CA19-9. Positive ctDNA results were associated with worse prognosis, including shorter OS and PFS, and may help guide treatment decisions. These findings suggest that ctDNA could be a valuable tool for personalized management in PDAC, though further prospective studies are needed to validate its clinical role in treatment stratification.
Collapse
Affiliation(s)
- Yuqi Zhang
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, TX 77094, USA
| | - Abdullah Esmail
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, TX 77094, USA
| | - Hala Hassanain
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, TX 77094, USA
| | - Vikramjit Dhillon
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, TX 77094, USA
| | - Waseem Abdelrahim
- Michael E. DeBakey HS for Health Professions, Houston, TX 77030, USA
| | - Ebtesam Al-Najjar
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, TX 77094, USA
| | - Bayan Khasawneh
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, TX 77094, USA
| | - Maen Abdelrahim
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, TX 77094, USA
| |
Collapse
|
48
|
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
|
49
|
Gonzalo-Encabo P, Gardiner J, Norris MK, Wilson RL, Normann AJ, Nguyen D, Parker N, Tjogas D, Brais LK, Meyerhardt JA, Rosenthal MH, Wolpin BM, Uno H, Dieli-Conwright CM. Resistance exercise combined with protein supplementation for skeletal muscle mass in people with pancreatic cancer undergoing neoadjuvant chemotherapy: Study protocol for the REBUILD trial. PLoS One 2025; 20:e0322192. [PMID: 40315221 PMCID: PMC12047797 DOI: 10.1371/journal.pone.0322192] [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: 08/26/2024] [Accepted: 03/10/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Pancreatic cancer patients' prognosis may be limited by two conditions, cachexia and sarcopenia. Resistance exercise and protein supplementation are safe non-pharmacological strategies that may increase or preserve skeletal muscle mass within this population. Therefore, the primary aim of this study is to examine the feasibility of a home-based virtually supervised resistance exercise intervention, with or without protein supplementation in pancreatic cancer patients initiating neoadjuvant chemotherapy. This intervention may also maintain skeletal muscle mass and improve plasma biomarkers associated with muscle tissue wasting, physical function and psychological measures. METHODS We aim to recruit 45 patients with locally advanced pancreatic cancer initiating neoadjuvant chemotherapy. Patients will be randomized to receive either Resistance Exercise (RE) (n = 15), Resistance Exercise and Protein Supplementation (RE + PS) (n = 15), or Attention Control (AC) (n = 15). Patients randomized to RE or RE + PS will receive 16-weeks of home-based virtually supervised resistance exercise. The AC will receive a 16-week stretching program. Primary and secondary outcomes will be measured at baseline and after 16 weeks during study visits. DISCUSSION The REBUILD trial is the first randomized controlled trial that combines resistance exercise with daily protein supplementation during neoadjuvant chemotherapy in pancreatic cancer patients. Our novel home-based virtually supervised exercise intervention seeks to mitigate barriers to participation in this vulnerable population. Furthermore, results of this trial will address important research gaps associated with pancreatic cancer-related cachexia, a condition closely connected with poor prognosis and mortality.
Collapse
Affiliation(s)
- Paola Gonzalo-Encabo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Departamento de Ciencias Biomédicas, Área de Educación Física y Deportiva, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Madrid, España
| | - John Gardiner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Mary K. Norris
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Rebekah L. Wilson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Amber J. Normann
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Health Sciences, Boston University, Boston, Massachusetts, United States of America
| | - Danny Nguyen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Nathan Parker
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Darryl Tjogas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Lauren K. Brais
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jeffrey A. Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Michael H. Rosenthal
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Imaging, Dana-Farber Cancer Institute, Boston Massachusetts, United States of America
- Department of Radiology, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Brian M. Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hajime Uno
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Christina M. Dieli-Conwright
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
50
|
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
|