1
|
Szász Z, Takács A, Kalabay M, Bárány P, Czuczi T, Csámpai A, Lajkó E, Kőhidai L. Comparative study of the anti-tumour effects of the imipridone, ONC201 and its fluorinated analogues on pancreatic cancer cell line. Sci Rep 2025; 15:15925. [PMID: 40335552 PMCID: PMC12059162 DOI: 10.1038/s41598-025-00070-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 04/24/2025] [Indexed: 05/09/2025] Open
Abstract
Pancreatic ductal adenocarcinoma has a high mortality rate, with a 5-year survival rate of ~ 12%. Therefore, developing new targeted therapies is urgently needed. ONC-201, a promising candidate, is currently undergoing clinical trials. The main objective of the present work is to investigate the anti-tumour activity of ONC-201 and its two fluorinated analogues (TBP-134, TBP-135). The viability of two pancreatic adenocarcinoma cell lines (PANC-1, MIA PaCa-2) and three other tumour cell lines (A2058, EBC-1, COLO-205) was assessed after 72-hour treatment with drugs at 0.5, 10, and 25 µM. Significant antiproliferative effects were observed, with 0.5 µM TBP-134 achieving the highest potency, reducing cell viability to approximately 50%. None of the molecules exhibited significant cytotoxicity toward normal human dermal fibroblast cells or cardiomyocytes, indicating a selective anti-tumour profile. The analogues showed more effective results than ONC201 on PANC-1 cells (IC50: 0.35 and 1.8 µM vs. IC50: 6.1 µM, respectively). All analogues induced G2/M phase arrest followed by apoptosis in PANC-1 cells. The site of the fluorination influenced the mechanism of apoptotic action of these compounds. Overall, TBP-134 showed superior efficacy, making it a promising candidate for structural optimization within the imipridone family to develop more effective, selective treatments for pancreatic tumours.
Collapse
Affiliation(s)
- Zsófia Szász
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, 1089, Hungary.
| | - Angéla Takács
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, 1089, Hungary
| | - Márton Kalabay
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, 1089, Hungary
| | - Péter Bárány
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, Budapest, 1117, Hungary
| | - Tamás Czuczi
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, Budapest, 1117, Hungary
| | - Antal Csámpai
- Department of Organic Chemistry, Institute of Chemistry, Eötvös Loránd University, Budapest, 1117, Hungary
| | - Eszter Lajkó
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, 1089, Hungary
| | - László Kőhidai
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, 1089, Hungary
| |
Collapse
|
2
|
Yadav K, Das T, Lynn AM. Pancancer analysis of DNA damage repair gene mutations and their impact on immune regulatory gene expression. Sci Rep 2025; 15:15667. [PMID: 40325163 PMCID: PMC12052996 DOI: 10.1038/s41598-025-99965-y] [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: 02/13/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025] Open
Abstract
DNA damage is a key factor in many human disorders, including cancer, chronic inflammation, and early aging. Genes involved in DNA damage repair (DDR) not only maintain genomic stability but also contribute to immune responses by regulating the expression of antimicrobial peptides and ligands that activate immune receptors. Various components of the DNA damage response (DDR), including DNA damage sensors, transducer kinases, and effector proteins, are capable of activating diverse immunological signaling pathways. While DDR gene mutations are common in cancers, their effects on immune characteristics are not well understood. We investigated how mutations in DDR genes influence the expression of immune regulatory genes, including immune stimulators, inhibitors, and genes related to the major histocompatibility complex (MHC) pathway. Using gene expression data from The Cancer Genome Atlas (TCGA) and mutation data from cBioPortal, we analyzed 264 DDR-related genes and 66 immune regulatory genes. These genes were clustered and categorized using Metascape, an integrative bioinformatics tool that applies enrichment-based analysis to group functionally related genes into clusters. The clustered genes were further validated through a literature review and the GeneCards database. We scored the change in immune regulatory gene expression in response to DDR gene mutations to identify differentially expressed immune stimulators, inhibitors, and MHC-related genes. Our analysis revealed positive and negative correlations between DDR gene mutations and the expression of immune modulators. These findings could help guide future cancer treatments based on biomarkers and immunotherapy strategies.
Collapse
Affiliation(s)
- Kanchana Yadav
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Trishala Das
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Andrew M Lynn
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
3
|
Nasti A, Inagaki S, Ho TTB, Seki A, Yoshida K, Satomura K, Sakai Y, Kaneko S, Yamashita T. Cystatin A promotes the antitumor activity of T helper type 1 cells and dendritic cells in murine models of pancreatic cancer. Mol Oncol 2025; 19:1452-1470. [PMID: 39792573 PMCID: PMC12077287 DOI: 10.1002/1878-0261.13796] [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: 05/29/2024] [Revised: 11/21/2024] [Accepted: 12/12/2024] [Indexed: 01/12/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a disease with poor prognosis due to diagnostic and therapeutic limitations. We previously identified cystatin A (CSTA) as a PDAC biomarker and have conducted the present study to investigate the antitumor effects of CSTA. PDAC murine models were established with genetically modified PAN02 tumor cell lines to evaluate the antitumor immune response. PDAC mouse survival was significantly longer with CSTA, and its antitumor effect was mediated mainly by CD4+ cells and partly by CD8+ cells. We also observed an increased infiltration of CD4+ and CD8+ cells in tumors of mice overexpressing CSTA. Phenotypically, we confirmed higher T helper type 1 (Th1) cell activity and increased frequency and activity of M1 macrophages and dendritic cells (DCs) in CSTA-overexpressing mice. Gene expression analysis highlighted pathways related to interferon gamma (IFN-γ) induction and Th1 lymphocyte activation that were induced by CSTA. Macrophages and DCs shifted toward proinflammatory antitumor phenotypes. Furthermore, activated splenocytes of PDAC model mice expressing CSTA had increased proapoptotic activity. CSTA also promoted the selective migration of CD4+ and CD11c+ immune cells in an in vitro migration assay. In conclusion, CSTA exerts antitumor effects by enhancing Th1-mediated antitumor effects through promotion of DC and M1 macrophage activity, thereby increasing immune cell chemotaxis. CSTA could be a novel therapeutic candidate for PDAC.
Collapse
Affiliation(s)
- Alessandro Nasti
- Information‐Based Medicine DevelopmentGraduate School of Medical Sciences, Kanazawa UniversityJapan
| | - Shingo Inagaki
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Tuyen Thuy Bich Ho
- Information‐Based Medicine DevelopmentGraduate School of Medical Sciences, Kanazawa UniversityJapan
| | - Akihiro Seki
- Department of GastroenterologyKanazawa University HospitalJapan
| | - Keiko Yoshida
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Kosuke Satomura
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Yoshio Sakai
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Shuichi Kaneko
- Information‐Based Medicine DevelopmentGraduate School of Medical Sciences, Kanazawa UniversityJapan
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
- Department of GastroenterologyKanazawa University HospitalJapan
| | - Taro Yamashita
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
- Department of GastroenterologyKanazawa University HospitalJapan
| |
Collapse
|
4
|
Naseem S, Sajid R, Nabeel M, Sadiqa A, Rizwan M, Zulfiqar MR, Ahmad A, Iqbal DN. Advancing nanocellulose-based biosensors: pioneering eco-friendly solutions for biomedical applications and sustainable material replacement. Int J Biol Macromol 2025; 309:143057. [PMID: 40220829 DOI: 10.1016/j.ijbiomac.2025.143057] [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: 01/30/2025] [Revised: 03/30/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
The escalating demand for sustainable and high-performance biosensing technologies has intensified interest in nanocellulose-based biosensors as eco-friendly alternatives to conventional materials. Nanocellulose, derived from abundant natural sources, offers remarkable properties such as high surface area, mechanical strength, biocompatibility, and chemical versatility, making it highly suitable for biosensing applications. This review delves into the synthesis, functionalization, and diverse applications of nanocellulose materials, particularly bacterial nanocellulose (BNC) and cellulose nanofibrils (CNFs), in the development of advanced biosensors. Innovative functionalization techniques, including polymer grafting and TEMPO oxidation, have been employed to enhance the specificity, stability, and sensitivity of these biosensors. These advancements lay the foundation for a sustainable and efficient biosensing framework, positioning nanocellulose-based technologies at the forefront of developing eco-friendly and accessible biosensors for biomedical applications and beyond.
Collapse
Affiliation(s)
- Sobia Naseem
- Department of Chemistry, University of Engineering and Technology, Lahore, Pakistan; Department of Polymer and Process Engineering, University of Engineering and Technology, Lahore, Pakistan
| | - Rumana Sajid
- Department of Chemistry, The University of Lahore, Lahore 54590, Pakistan
| | - Muhammad Nabeel
- Department of Chemistry, The University of Lahore, Lahore 54590, Pakistan
| | - Ayesha Sadiqa
- Department of Chemistry, The University of Lahore, Lahore 54590, Pakistan.
| | - Muhammad Rizwan
- Department of Chemistry, University of Engineering and Technology, Lahore, Pakistan.
| | | | - Awais Ahmad
- Department of Chemistry, The University of Lahore, Lahore 54590, Pakistan.
| | - Dure Najaf Iqbal
- Department of Chemistry, The University of Lahore, Lahore 54590, Pakistan
| |
Collapse
|
5
|
Pele KG, Calderón-Villalba A, Amaveda H, Mora M, Zhang-Zhou J, Pérez MÁ, García-Aznar JM, Alamán-Díez P, García-Gareta E. Novel hydrogel-based cancer-on-a-chip models for growth of 3D multi-cellular structures and investigation of early angiogenesis in pancreatic ductal adenocarcinoma. Colloids Surf B Biointerfaces 2025; 253:114736. [PMID: 40315572 DOI: 10.1016/j.colsurfb.2025.114736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/27/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
Cancer-on-a-chip models have enormous potential for the study of tumour development events. Here, we investigated hydrogels of egg white (EW) and gelatin for growth of 3D multi-cellular structures and investigation of early angiogenesis inside microfluidic devices. We focused on pancreatic ductal adenocarcinoma (PDAC), a devastating gastrointestinal malignancy. EW/gelatin hydrogels were stiffer and showed porous globular structures compared to the fibrous network of collagen I molecules. PANC-1 cells preferentially formed significantly larger spheroids in collagen I than in EW/gelatin hydrogels, whilst cell aggregates in the shape of grape-like clusters were significantly larger and more abundant in EW/gelatin. Cells inside the aggregates showed active cell unions, secreted matrix, and formed active unions with the surrounding EW/gelatin hydrogel. Early stages of PDAC were recreated by co-culture of two different microenvironments, one for PANC-1 and another one for fibroblasts, for investigating the secretion of soluble angiogenic factors, which depended on the role of each factor in the angiogenic and tumorigenic processes. Overall, cancer cell proliferation and establishment of a tumour vasculature were favoured. This study demonstrates the importance of the microenvironment in tumour cells behaviour as well as the complex interplay between the different cells present in PDAC to establish a tumoural vasculature.
Collapse
Affiliation(s)
- Karinna Georgiana Pele
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Alejandro Calderón-Villalba
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Hippolyte Amaveda
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC and University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Mario Mora
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC and University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Jack Zhang-Zhou
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - María Ángeles Pérez
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain
| | - José Manuel García-Aznar
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain
| | - Pilar Alamán-Díez
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Elena García-Gareta
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain; Division of Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, University College London, London NW3 2QG, United Kingdom.
| |
Collapse
|
6
|
Kyuno D, Asano H, Okumura R, Takasawa K, Takasawa A, Konno T, Nakamori Y, Magara K, Ono Y, Imamura M, Kimura Y, Kojima T, Osanai M. The Role of Claudin-1 in Enhancing Pancreatic Cancer Aggressiveness and Drug Resistance via Metabolic Pathway Modulation. Cancers (Basel) 2025; 17:1469. [PMID: 40361399 PMCID: PMC12070999 DOI: 10.3390/cancers17091469] [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: 04/08/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pancreatic ductal adenocarcinoma is a lethal malignancy, necessitating an understanding of its molecular mechanisms for the development of new therapeutic strategies. The tight junction protein claudin-1, known to influence cellular functions in various cancers and is considered a therapeutic target, remains unclear in pancreatic cancer. METHODS This study assessed claudin-1 expression in resected pancreatic cancer samples, public databases, and pancreatic cancer cell lines. Claudin-1 knockout with CRISPR/Cas9 on poorly differentiated pancreatic cancer cell lines and a proteome analysis were performed to investigate the intracellular mechanisms of claudin-1. RESULTS Claudin-1 was markedly overexpressed in pancreatic ductal adenocarcinoma and intraepithelial neoplasia compared to normal ducts, and high claudin-1 levels were an independent predictor of poor prognosis. Claudin-1 knockout diminished cell proliferation, migration, invasion, and chemoresistance in pancreatic ductal adenocarcinoma. Proteome analysis revealed the significant downregulation of aldo-keto reductase family proteins (AKR1C2, AKR1C3, and AKR1B1) in claudin-1 knockout cells, which are linked to metabolic pathways. Aldo-keto reductase knockdown reduced chemoresistance, proliferation, and invasion in these cell lines. CONCLUSIONS These findings indicate that the abnormal expression of claudin-1 promotes tumor progression and drug resistance through its interaction with aldo-keto reductase proteins, highlighting claudin-1 and aldo-keto reductase family proteins as potential biomarkers and therapeutic targets for pancreatic cancer.
Collapse
Affiliation(s)
- Daisuke Kyuno
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
- Department of Surgery, Division of Gastroenterological Surgery, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Hinae Asano
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Reona Okumura
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Kumi Takasawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - Akira Takasawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | - Takumi Konno
- Department of Cell Science, Institute of Cancer Research, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Yuna Nakamori
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Kazufumi Magara
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Yusuke Ono
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Masafumi Imamura
- Department of Surgery, Division of Gastroenterological Surgery, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Yasutoshi Kimura
- Department of Surgery, Division of Gastroenterological Surgery, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Takashi Kojima
- Department of Cell Science, Institute of Cancer Research, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Makoto Osanai
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| |
Collapse
|
7
|
Lazzini G, Gaeta R, Pollina LE, Comandatore A, Furbetta N, Morelli L, D'Acunto M. Raman spectroscopy based diagnosis of pancreatic ductal adenocarcinoma. Sci Rep 2025; 15:13240. [PMID: 40247119 PMCID: PMC12006465 DOI: 10.1038/s41598-025-98122-9] [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: 11/28/2024] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Pancreatic ductal adenocarcinoma is currently the 12th most frequent form of cancer worldwide, characterized by a very low 5-year survival rate. Although several therapeutic approaches have been proposed to treat this form of pancreatic cancer, surgical resection is still commonly recognized as the most effective technique to slow down the disease progression and maximize the 5-year survival rate. Analogously, one critical issue is the ability of current diagnostic methodologies to distinguish between irregular growth of the tumor mass and surrounding inflammatory tissues. In this pilot study, we apply Raman spectroscopy, supported by a series of machine learning techniques, to distinguish among healthy, pancreatitis and ductal adenocarcinoma tissues, respectively, for a total of 15 cases. Raman spectroscopy is a label-free, non-destructive spectral technique exploiting Raman scattering. In turn, by applying a combination of principal component analysis and random forest classifier on the Raman spectral dataset, we achieved a maximum accuracy of up to ∼ 96%. Our findings clearly indicate that Raman spectroscopy could become a powerful spectral technique to support pathologists in improving pancreatic cancer diagnosis.
Collapse
Affiliation(s)
- Gianmarco Lazzini
- CNR-IBF, Istituto di Biofisica Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124, Pisa, Italy
| | - Raffele Gaeta
- Second Division of Surgical Pathology, University Hospital of Pisa, Pisa, Italy
| | | | - Annalisa Comandatore
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Department of Surgery, Amsterdam UMC, Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Niccolò Furbetta
- Department of Surgery, Amsterdam UMC, Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Luca Morelli
- Department of Surgery, Amsterdam UMC, Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Mario D'Acunto
- CNR-IBF, Istituto di Biofisica Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124, Pisa, Italy.
| |
Collapse
|
8
|
Stoop TF, Javed AA, Oba A, Koerkamp BG, Seufferlein T, Wilmink JW, Besselink MG. Pancreatic cancer. Lancet 2025; 405:1182-1202. [PMID: 40187844 DOI: 10.1016/s0140-6736(25)00261-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 04/07/2025]
Abstract
Pancreatic cancer is frequently a lethal disease with an aggressive tumour biology often presenting with non-specific symptoms. Median survival is approximately 4 months with a 5-year survival of 13%. Surveillance is recommended in individuals with familial pancreatic cancer, specific mutations, and high-risk intraductal papillary mucinous neoplasm, as they are at high risk of developing pancreatic cancer. Chemotherapy combined with surgical resection remains the cornerstone of treatment. However, only a small subset of patients are candidates for surgery. Multi-agent chemotherapy has improved survival in the palliative setting for patients with metastatic disease, as (neo)adjuvant and induction therapy have in patients with borderline resectable and locally advanced pancreatic. Given that pancreatic cancer is predicted to become the second leading cause of cancer-related death by 2030, novel therapies are urgently needed.
Collapse
Affiliation(s)
- Thomas F Stoop
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Ammar A Javed
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands; Division of Surgical Oncology, Department of Surgery, New York University Medical Center, New York, NY, USA
| | - Atsushi Oba
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan; Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Thomas Seufferlein
- Department of International Medicine I, Ulm University Hospital, Ulm, Germany
| | - Johanna W Wilmink
- Department of Medical Oncology, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Marc G Besselink
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
9
|
Gorbokon N, Teljuk K, Reiswich V, Lennartz M, Minner S, Simon R, Sauter G, Wilczak W, Clauditz TS, Schraps N, Hackert T, Uzunoglu FG, Kluth M, Bubendorf L, Matter M, Viehweger F, Freytag M, Jacobsen F, Möller K, Steurer S. Deficiency of MTAP Is Frequent and Mostly Homogeneous in Pancreatic Ductal Adenocarcinomas. Cancers (Basel) 2025; 17:1205. [PMID: 40227771 PMCID: PMC11987894 DOI: 10.3390/cancers17071205] [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: 02/03/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND The complete loss of S-methyl-5'-thioadenosine phosphorylase (MTAP) expression, often due to homozygous 9p21 deletion, creates a druggable vulnerability in cancer cells. METHODS A total of 769 primary pancreatic ductal adenocarcinomas were analyzed on tissue microarrays with MTAP immunohistochemistry (IHC) and 9p21 fluorescence in situ hybridization (FISH). Intratumoral heterogeneity was assessed on a "heterogeneity" TMA containing up to nine samples from different areas of 236 primary tumor and nodal metastases, and whole sections of all tumor blocks from 19 cancers. RESULTS MTAP expression loss was found in 181 (37.9%) of 478 interpretable primary tumors and was unrelated to pT, pN, grade, and tumor size. MTAP expression loss was homogenous in 37.6% and heterogeneous in 1.1% of the 181 tumors, with at least three evaluable samples on the heterogeneity TMA. On whole sections, 1 of 19 tumors showed heterogeneous MTAP loss. The correlation between IHC and FISH was nearly perfect, with 98.8% of MTAP-deficient samples showing a 9p21 deletion. CONCLUSIONS MTAP expression loss is frequent, caused by homozygous deletion, and mostly homogeneous in pancreatic ductal adenocarcinomas. Considering also their aggressive clinical behavior, pancreatic adenocarcinomas may represent an ideal cancer type for studying new drugs targeting MTAP-deficient cancer cells in clinical trials.
Collapse
Affiliation(s)
- Natalia Gorbokon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Katharina Teljuk
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Viktor Reiswich
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Waldemar Wilczak
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Till Sebastian Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Nina Schraps
- General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.S.); (T.H.); (F.G.U.)
| | - Thilo Hackert
- General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.S.); (T.H.); (F.G.U.)
| | - Faik G. Uzunoglu
- General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.S.); (T.H.); (F.G.U.)
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland; (L.B.); (M.M.)
| | - Matthias Matter
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland; (L.B.); (M.M.)
| | - Florian Viehweger
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Morton Freytag
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Frank Jacobsen
- Pathologie-Hamburg, Labor Lademannbogen MVZ GmbH, 22419 Hamburg, Germany;
| | - Katharina Möller
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| |
Collapse
|
10
|
Hill JLE, Leonard E, Parslow D, Hill DJ. Gene Dysregulation and Islet Changes in PDAC-Associated Type 3c Diabetes. Int J Mol Sci 2025; 26:3191. [PMID: 40244011 PMCID: PMC11988973 DOI: 10.3390/ijms26073191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy, often associated with new-onset diabetes. The relationship between PDAC and diabetes, particularly type 3c diabetes, remains poorly understood. This study investigates whether PDAC-associated diabetes represents a distinct subtype by integrating transcriptomic and histological analyses. Whole-tumour RNA sequencing data from The Cancer Genome Atlas (TCGA) were analysed to compare gene expression profiles between PDAC patients with and without diabetes. Cell-type Identification By Estimating Relative Subsets Of RNA Transcripts (CIBERSORT) deconvolution was employed to assess immune cell populations. Histopathological evaluations of pancreatic tissues were conducted to assess fibrosis and islet morphology. Histological analysis revealed perivascular fibrosis and islet basement membrane thickening in both PDAC cohorts. Transcriptomic data indicated significant downregulation of islet hormone genes insulin (INS) and glucagon (GCG) but not somatostatin (SST) in PDAC-associated diabetes, consistent with a type 3c diabetes phenotype. Contrary to previous reports, no distinct immunogenic signature was identified in PDAC with diabetes, as key immune checkpoint genes (Programmed Cell Death Protein 1 (PDCD1), Cytotoxic T-Lymphocyte Associated Protein 4 (CTLA4), Programmed Death-Ligand 1(PD-L1)) were not differentially expressed. The findings suggest that PDAC-associated diabetes arises through neoplastic alterations in islet physiology rather than immune-mediated mechanisms. The observed reductions in endocrine markers reinforce the concept of PDAC-driven β-cell dysfunction as a potential early indicator of malignancy. Given the poor response of PDAC to PD-L1 checkpoint inhibitors, further research is needed to elucidate alternative therapeutic strategies targeting tumour-islet interactions.
Collapse
Affiliation(s)
| | - Eliot Leonard
- Leeds Teaching Hospitals NHS Trust, Leeds LS1 3EX, UK;
| | | | - David J. Hill
- Lawson Research Institute, St. Joseph Health Care, London, ON N6A 4V2, Canada
- Departments of Medicine, Physiology and Pharmacology, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
11
|
Petrauskas V, Damaseviciute R, Gulla A. Pancreatic 3D Organoids and Microfluidic Systems-Applicability and Utilization in Surgery: A Literature Review. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:623. [PMID: 40282914 PMCID: PMC12028617 DOI: 10.3390/medicina61040623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/05/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025]
Abstract
Background: Pancreatic organoids are a rapidly advancing field of research with new discoveries being made every day. A literature review was performed to answer the question of how relevant 3D pancreatic organoids are for surgery. Materials and Methods: We started our investigation by identifying articles in PubMed within the last 5 years using the keywords (("pancreatic organoid", OR "organ-on-a-chip", OR "pancreatic chip" OR "3D culture methods") AND pancreatic surgery). Only English articles were included in this literature review. This literature review was performed in a non-systematic way; articles were chosen without a predetermined protocol of inclusion and were based on the aim of the review. Results and Conclusions: There are many promising innovations in the field of 3D cultures. Drug sensitivity testing in particular holds great potential for surgical application. For locally advanced PDAC, EUS-FNB obtained cancer tissue can be cultured as organoids, and after 4 weeks, neoadjuvant treatment could be adjusted for each patient individually. Utilizing this approach could increase the number of R0 resections and possibly cure the disease. Furthermore, microfluidic devices, as a platform for pancreatic islet pre-transplant evaluation or cultivation of beta cells derived from HiPSC in vitro, promise broad application of islet transplantation to T1DM patients in the near future.
Collapse
Affiliation(s)
- Vidas Petrauskas
- Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, LT-01131 Vilnius, Lithuania
| | - Ryte Damaseviciute
- Center of Visceral Medicine and Translational Research, Faculty of Medicine, Vilnius University, LT-01131 Vilnius, Lithuania
| | - Aiste Gulla
- Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, LT-01131 Vilnius, Lithuania
- Center of Visceral Medicine and Translational Research, Faculty of Medicine, Vilnius University, LT-01131 Vilnius, Lithuania
- Department of Surgery, George Washington University, Washington, DC 20052, USA
| |
Collapse
|
12
|
Willink CY, Jenniskens SFM, Klaassen NJM, Stommel MWJ, van Laarhoven CJHM, Fütterer JJ, Nijsen JFW. Development of an Intratumoral Holmium Microsphere Injection Method in Ex Vivo Human Pancreatic Ductal Adenocarcinoma: A Preclinical Feasibility Study. Cancers (Basel) 2025; 17:1028. [PMID: 40149361 PMCID: PMC11941289 DOI: 10.3390/cancers17061028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Patients diagnosed with pancreatic ductal adenocarcinoma (PDAC) have a poor prognosis. Local therapy may enhance tumor control and increase resectability. Intratumoral injection of radioactive holmium-166 microspheres presents a promising and minimally invasive treatment with multimodality imaging capabilities (SPECT, CT, MRI). However, holmium-166 microspheres are not commonly used for intratumoral injections, and PDAC is notorious for its high intratumoral pressure. This study developed an intratumoral injection method with nonradioactive holmium-165 microspheres in ex vivo human PDAC specimens using a novel injection system for suspension homogenization. METHODS An injection system was developed and validated in a laboratory setting. Thereafter, intratumoral injections in surgically removed ex vivo PDACs were performed, and parameters were established to optimize feasibility, defined by the ability to inject and control the microsphere distribution. Also, injection limitations and cutoff values were determined. The distribution was assessed by visual confirmation, CT, MRI, ultrasound, and histopathology. RESULTS With a validated injection system, intratumoral injections were performed in ten ex vivo PDAC samples. Feasible injection guidelines include but are not limited to ultrasound or CT needle guidance, a maximum injection volume of <20.0% from the tumor volume, ≤3 needle positions, and an injection volume of 0.3-1.0 mL per needle position. CONCLUSIONS Intratumoral injection of holmium-165 microspheres in ex vivo pancreatic ductal adenocarcinoma was feasible with adherence to injection parameters necessary for effective intratumoral deposition and minimal leakage. The injection system and parameters developed here provide a foundation for future studies on holmium-166 microsphere injections in pancreatic cancer patients, with the aim to improve local tumor control as a part of a multimodal therapy.
Collapse
Affiliation(s)
- Coen Ysbrand Willink
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein-Zuid 10, Postbox 9101, 6500 HB Nijmegen, The Netherlands (J.F.W.N.)
| | - Sjoerd Franciscus Maria Jenniskens
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein-Zuid 10, Postbox 9101, 6500 HB Nijmegen, The Netherlands (J.F.W.N.)
| | - Nienke Johanna Maria Klaassen
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein-Zuid 10, Postbox 9101, 6500 HB Nijmegen, The Netherlands (J.F.W.N.)
| | - Martijn Willem Jan Stommel
- Department of Surgery, Radboud University Medical Center, Geert Grooteplein-Zuid 10, Postbox 9101, 6500 HB Nijmegen, The Netherlands
| | | | - Jurgen J. Fütterer
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein-Zuid 10, Postbox 9101, 6500 HB Nijmegen, The Netherlands (J.F.W.N.)
| | - Johannes Frank Wilhelmus Nijsen
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein-Zuid 10, Postbox 9101, 6500 HB Nijmegen, The Netherlands (J.F.W.N.)
| |
Collapse
|
13
|
Cho CJ, Nguyen T, Rougeau AK, Huang YZ, To S, Lin X, Thalalla Gamage S, Meier JL, Mills JC. Inhibition of Ribosome Biogenesis In Vivo Causes p53-Dependent Death and p53-Independent Dysfunction. Cell Mol Gastroenterol Hepatol 2025; 19:101496. [PMID: 40081569 DOI: 10.1016/j.jcmgh.2025.101496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND & AIMS Although it is well-known that ribosomes are critical for cell function, and their synthesis (known as ribosome biogenesis [RiBi]) is energy-intensive, surprisingly little is known about RiBi in vivo in adult tissue. METHODS Using a mouse model with conditional deletion of Nat10, an essential gene for RiBi and subsequent translation of mRNA, we investigated the effects of RiBi blockade in vivo, with a focus on pancreatic acinar cells during homeostasis and tumorigenesis. RESULTS We observed an unexpected latency of several weeks between Nat10 deletion and onset of structural and functional abnormalities and p53-dependent acinar cell death. Although deletion of Trp53 rescued acinar cells from apoptotic cell death, Nat10Δ/Δ; Trp53Δ/Δ acinar cells remained morphologically and functionally abnormal. Deletion of Nat10 in acinar cells blocked Kras-oncogene-driven pancreatic ductal adenocarcinoma, regardless of Trp53 mutation status. CONCLUSIONS Together, our results provide initial insights into how differentiated cells respond to defects in RiBi and translation in vivo in various physiological contexts.
Collapse
Affiliation(s)
- Charles J Cho
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas.
| | - Thanh Nguyen
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Amala K Rougeau
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Yang-Zhe Huang
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Sarah To
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Xiaobo Lin
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Supuni Thalalla Gamage
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Jordan L Meier
- Chemical Biology Laboratory, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Jason C Mills
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas; Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
14
|
Wen Y, Zhao G, Dai C. Cell-free DNA: plays an essential role in early diagnosis and immunotherapy of pancreatic cancer. Front Immunol 2025; 16:1546332. [PMID: 40124355 PMCID: PMC11925872 DOI: 10.3389/fimmu.2025.1546332] [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] [Accepted: 02/24/2025] [Indexed: 03/25/2025] Open
Abstract
Pancreatic cancer is renowned for its aggressive nature and dismal prognosis, with the majority of patients diagnosed at an advanced stage. The prognosis for patients with pancreatic cancer can be improved by early diagnosis and effective treatment. Circulating cell-free DNA (cfDNA) has emerged as a promising biomarker for the early diagnosis and monitoring of pancreatic cancer. This research presents a review of circulating cell-free DNA essential role in the early diagnosis and immunotherapy of pancreatic cancer. The detection methods of cfDNA, its potential as a diagnostic biomarker, and the latest research progress in cfDNA-based immunotherapy are discussed. The findings suggest that cfDNA plays a vital role in the early detection and personalised treatment of pancreatic cancer, holding great promise for improving patient outcomes.
Collapse
Affiliation(s)
- Yi Wen
- College of Outstanding Clinician, Jiangsu University, Zhenjiang, China
| | - Gengmin Zhao
- Graduate School, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Chunhua Dai
- Department of Thoracic Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
15
|
Lockie EB, Sylivris A, Pandanaboyana S, Zalcberg J, Skandarajah A, Loveday BP. Relationship between pancreatic cancer resection rate and survival at population level: systematic review. BJS Open 2025; 9:zraf007. [PMID: 40131791 PMCID: PMC11934921 DOI: 10.1093/bjsopen/zraf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/17/2024] [Accepted: 01/10/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Surgery combined with chemotherapy provides the best chance of survival in pancreatic cancer. This study investigated whether increasing the resection rate at a population level improves overall survival and modelled the interaction between resection rate, perioperative mortality rate, and population survival. METHODS A systematic review was conducted on studies reporting resection rate and survival outcomes in patients with pancreatic cancer at a population level. MEDLINE, Embase and Evidence-Based Medicine Reviews were searched up to February 2024. The primary outcome was overall population-level survival. A model for 1-year survival incorporating varying resection and perioperative mortality rates was developed. RESULTS The search identified 3967 studies; 19 were eligible (516 789 patients). A significant association was observed between resection rate and pancreatic cancer population survival at 1 year (r2 = 0.46, P = 0.001). A weak but significant association was noted between resection rate and (neo)adjuvant chemotherapy (r2 = 0.26, P = 0.03). One-year pancreatic cancer population survival was significantly associated with chemotherapy (r2 = 0.63; P = 0.004), but the effect was weaker than for resection rate (regression slope 0.26 versus 0.94 respectively). According to the developed model, for example, increasing the resection rate from 10 to 15% and perioperative mortality rate from 2 to 3% would lead to a 1-year survival increase from 17.6% to 22.1%. CONCLUSION A higher resection rate at a population level was associated with improved survival of the pancreatic cancer population. While some of this benefit was linked to increasing (neo)adjuvant chemotherapy use, the effect of resection rate was stronger. Strategies to enhance the resection rate at national and regional levels should be explored. Establishing a benchmark for resection rate could support patient-centred healthcare and promote equitable access to high-quality pancreatic cancer care.
Collapse
Affiliation(s)
- Elizabeth B Lockie
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of General Surgical Specialties, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Amy Sylivris
- Department of General Surgical Specialties, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Sanjay Pandanaboyana
- Hepato-Pancreatico-Biliary Centre, Freeman Hospital, Newcastle upon Tyne, UK
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - John Zalcberg
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- School of Public Health, Faculty of Medicine, Monash University, Clayton, Victoria, Australia
- Department of Medical Oncology, Alfred Health, South Yarra, Victoria, Australia
| | - Anita Skandarajah
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of General Surgical Specialties, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Surgery, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
| | - Benjamin P Loveday
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of General Surgical Specialties, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Surgery, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia
| |
Collapse
|
16
|
Al Subeh Z, Pierre HC, Pearce CJ, Grinstaff MW, Colby AH, Liu K, Oberlies NH. Verticillin A-Loaded Surgical Buttresses Prevent Local Pancreatic Cancer Recurrence in a Murine Model. Mol Pharm 2025; 22:1220-1229. [PMID: 39868439 PMCID: PMC11881038 DOI: 10.1021/acs.molpharmaceut.4c00589] [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: 05/28/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/28/2025]
Abstract
The fungal metabolite verticillin A is a potent and selective histone methyltransferase inhibitor. It regulates apoptosis, the cell cycle, and stress response, and displays potent activity in the suppression of tumor cell growth in several different in vivo models. Verticillin A sensitizes pancreatic cancer cells to anti-PD-1 immunotherapy by regulating PD-L1 expression. However, as with many natural products, delivery and systemic toxicity are challenges that must be overcome to advance their use as a chemotherapeutic. To both reduce systemic toxicity and improve delivery, we report a verticillin A-loaded surgical buttress, which is well-tolerated at a dose as high as 40 mg/kg. In contrast, free verticillin A administered systemically results in toxicity at a dose of 3 mg/kg. The verticillin A-loaded buttress suppresses tumor recurrence in vivo in a safe and dose-dependent manner against a highly aggressive and metastatic model of pancreatic cancer.
Collapse
Affiliation(s)
- Zeinab
Y. Al Subeh
- Department
of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
- Department
of Biochemistry and Molecular Biology, Medical
College of Georgia, Augusta, Georgia 30912, United States
| | - Herma C. Pierre
- Department
of Chemistry and Biochemistry, University
of North Carolina at Greensboro, Greensboro, North Carolina 27402, United States
| | - Cedric J. Pearce
- Mycosynthetix,
Inc., Hillsborough, North Carolina 27278, United States
| | - Mark W. Grinstaff
- Departments
of Biomedical Engineering and Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Aaron H. Colby
- Departments
of Biomedical Engineering and Chemistry, Boston University, Boston, Massachusetts 02215, United States
- Ionic Pharmaceuticals,
LLC, Watertown, Massachusetts 02472, United States
| | - Kebin Liu
- Department
of Biochemistry and Molecular Biology, Medical
College of Georgia, Augusta, Georgia 30912, United States
- Georgia Cancer
Center, Medical College of Georgia, Augusta, Georgia 30912, United States
- Charlie
Norwood VA Medical Center, Augusta, Georgia 30904, United States
| | - Nicholas H. Oberlies
- Department
of Chemistry and Biochemistry, University
of North Carolina at Greensboro, Greensboro, North Carolina 27402, United States
| |
Collapse
|
17
|
Rahimian S, Mirkazemi K, Kamalinejad A, Doroudian M. Exosome-based advances in pancreatic cancer: The potential of mesenchymal stem cells. Crit Rev Oncol Hematol 2025; 207:104594. [PMID: 39732301 DOI: 10.1016/j.critrevonc.2024.104594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/28/2024] [Accepted: 12/08/2024] [Indexed: 12/30/2024] Open
Abstract
Pancreatic cancer, especially pancreatic ductal adenocarcinoma (PDAC), is one of the most challenging clinical conditions due to its late-stage diagnosis and poor survival rates. Mesenchymal stem cells (MSCs), used for targeted therapies, are being explored as a promising treatment because of their tumor-homing properties and potential contributions to the pancreatic cancer microenvironment. Understanding these interactions is crucial for developing effective treatments. In this study, we investigated how MSCs exhibit tropism towards tumors, influence the microenvironment through paracrine effects, and serve as potential drug delivery vehicles. We also examined their role in progression and therapeutic resistance in pancreatic cancer therapy. The cytotoxic effects of certain compounds on tumor cells, the use of genetically modified MSCs as drug carriers, and the potential of exosomal biomarkers like miRNAs and riRNAs for diagnosis and monitoring of pancreatic cancer were analyzed. Overall, MSC-based therapies, coupled with insights into tumor-stromal interactions, offer new avenues for improving outcomes in pancreatic cancer treatment. Additionally, the use of MSC-based therapies in clinical trials is discussed. While MSCs show promising potential for pancreatic cancer monitoring, diagnosis, and treatment, results so far have been limited.
Collapse
Affiliation(s)
- Sana Rahimian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Kimia Mirkazemi
- Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Armita Kamalinejad
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mohammad Doroudian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| |
Collapse
|
18
|
Chun JW, Shon HW, Gong JE, Lee MR, Lee YS, Kim SJ, Kang S, Kim S, Lee KY, Woo SM, Cho IR, Paik WH, Lee WJ, Kong SY, Ryu JK, Kim YT, Lee SH, Kim YH. ATR inhibition promotes synergistic antitumor effect in platinum-resistant pancreatic cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167625. [PMID: 39689762 DOI: 10.1016/j.bbadis.2024.167625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Oxaliplatin is a commonly used platinum-based chemotherapy drug for patients with pancreatic cancer (PC). Drug resistance is a major challenge in PC treatment, underscoring the urgent need for new approaches. Targeting DNA damage repair, one of the factors responsible for platinum resistance, is an attractive strategy to overcome drug resistance. This study aimed to investigate the potential of the ATR inhibitor BAY 1895344 in improving the drug responsiveness of oxaliplatin-resistant PC. METHODS Oxaliplatin-resistant PC cells (CFPAC-1 and Capan-2) were selected and treated with oxaliplatin, BAY 1895344, or a combination of both in vivo and in vitro. Their combinatorial effects on the DNA damage response (DDR) signaling pathway, apoptosis, and extent of DNA damage were evaluated using appropriate methods. Patient response was predicted using organoid models. RESULTS Combination treatment with BAY 1895344 and oxaliplatin exhibited a synergistic effect on both PC cell lines, with the effect being more pronounced on Capan-2. Additionally, the combination treatment substantially suppressed phospho-Chk1, a coordinator of DDR and cell cycle checkpoints. Mechanistically, ATR inhibition augmented the DNA damage induced by oxaliplatin, leading to mitotic catastrophe and cell death. Furthermore, in an in vivo study using a tumor-bearing xenograft mouse model, the combination treatment markedly reduced tumor growth. This synergistic effect was confirmed in patient-derived organoids with poor response to oxaliplatin. CONCLUSION ATR inhibition enhanced the anticancer effect of oxaliplatin, suggesting that this combination treatment could be an effective therapeutic strategy for overcoming platinum resistance in PC.
Collapse
Affiliation(s)
- Jung Won Chun
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea; Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea; Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Hye Won Shon
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Jeong Eun Gong
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Mi Rim Lee
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Yu-Sun Lee
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Sung Joon Kim
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Sumin Kang
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea; Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Sunshin Kim
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Kyung Yong Lee
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea; Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Sang Myung Woo
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea; Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea; Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - In Rae Cho
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Woo Hyun Paik
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Woo Jin Lee
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea; Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Sun-Young Kong
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea; Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Ji Kon Ryu
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Yong-Tae Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Sang Hyub Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea.
| | - Yun-Hee Kim
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea; Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Republic of Korea.
| |
Collapse
|
19
|
Kwon JY, Vera RE, Fernandez-Zapico ME. The multi-faceted roles of cancer-associated fibroblasts in pancreatic cancer. Cell Signal 2025; 127:111584. [PMID: 39756502 PMCID: PMC11807759 DOI: 10.1016/j.cellsig.2024.111584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/13/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025]
Abstract
The tumor microenvironment (TME) has been linked with the pathogenesis of pancreatic ductal adenocarcinoma (PDAC), the most common histological subtype of pancreatic cancer. A central component of the TME are cancer-associated fibroblasts (CAFs), which can either suppress or promote tumor growth in a context-dependent manner. In this review, we will discuss the multi-faceted roles of CAFs in tumor-stroma interactions influencing cancer initiation, progression and therapeutic response.
Collapse
Affiliation(s)
- John Y Kwon
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN 55901, USA.
| | - Renzo E Vera
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Rochester, MN 55901, USA.
| | | |
Collapse
|
20
|
Galouzis N, Khawam M, Alexander EV, Mesropyan L, Luu C, Khreiss MR, Riall TS. Quality of life and social health in patients after pancreatic surgery. J Gastrointest Surg 2025; 29:101969. [PMID: 39848313 DOI: 10.1016/j.gassur.2025.101969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Clinicians lack robust data on quality of life (QOL) and social functioning after pancreatectomy limiting their ability guide patient decision making aligned with patients' goals of care. METHODS In this cross-sectional survey study, we administered the European Organization for Research and Treatment of Cancer Core Quality of Life questionnaire, pancreas-specific QLQ-PAN26, Patient-Reported Outcomes Measurement Information System (PROMIS) Ability to Participate in Social Roles, and PROMIS activities and social isolation scales to all elective pancreatectomies (2021-2023). Results were compared with both normative data and between groups to determine factors predicting better QOL with a >10-12-point change considered clinically significant. RESULTS A total of 143 patients were included; 71 (49.6%) completed the distributed surveys. The average age of responders was 59.9 ± 16.1 years with 56.3% men. Pancreaticoduodenectomy (54.9%) was performed for malignancy in 67.6% of cases. Compared with normative population controls, postpancreatectomy patients reported lower role functioning scores (67.2 ± 28.7 vs 81.7 ± 28.2, 14.5 score difference) but less social isolation (40.6 ± 5.7 vs 50.0 ± 10.0, 9.4 score difference). Compared with patients with benign disease, those with malignancy reported clinically significant worse social functioning; more fatigue, pain, constipation, change in taste, weight loss, weakness, and altered bowel habits; worse body image; and increased worries about the future. Despite more symptoms, they were more satisfied with the healthcare they received (all >10-point score difference). CONCLUSION QOL and social health are affected by pancreatic resection and outcomes differ whether surgery was performed for benign or malignant disease. These issues are largely unaddressed and are potential targets for intervention to improve QOL.
Collapse
Affiliation(s)
- Nicholas Galouzis
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| | - Maria Khawam
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| | | | - Lusine Mesropyan
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| | - Carrie Luu
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| | | | - Taylor S Riall
- Department of Surgery, University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
21
|
Kikuyama M, Nakahodo J, Chiba K, Honda G. Focal pancreatic parenchymal atrophy could be a precursor of pancreatic cancer. Pancreatology 2025; 25:241-249. [PMID: 39894733 DOI: 10.1016/j.pan.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 12/10/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND/OBJECTIVES We previously reported that focal pancreatic parenchymal atrophy (FPPA) indicates high-grade pancreatic intraepithelial neoplasia (HG-PanIN) or carcinoma in situ (CIS). Because HG-PanIN progresses into pancreatic ductal adenocarcinoma (PDAC), the relationship between FPPA and PDAC should be investigated. METHODS We included 54 patients with PDAC, whose previous computed tomography or magnetic resonance imaging were reviewed. The existence, positional relationship between FPPA and PDAC, and time between FPPA recognition and PDAC diagnosis were all examined. Of the 54 patients, 28 underwent surgery. The remaining 26 patients were histopathologically diagnosed with PDAC using endoscopic ultrasonography-guided fine needle aspiration. RESULTS Among the 54 patients included, 49 (83.3 %) had FPPA. The pancreatic head and body were the common sites of FPPA. In all patients with FPPA, PDAC developed near the FPPA, with an average distance of 7.93 mm between the edge of the FPPA and the center of the PDAC. The interval between FPPA recognition and PDAC diagnosis was 35.33 months, which was significantly shorter in the surgical group. CONCLUSIONS FPPA could be a precursor of PDAC and suggest the area at risk of PDAC.
Collapse
Affiliation(s)
- Masataka Kikuyama
- Department of Gastroenterology, Tokyo Women's Medical University Hospital, Tokyo, Japan.
| | - Jun Nakahodo
- Department of Gastroenterology, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | - Kazuro Chiba
- Department of Gastroenterology, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | - Goro Honda
- Department of Surgery, Tokyo Women's Medical University Hospital, Tokyo, Japan
| |
Collapse
|
22
|
Patel A, Lancellotti F, Siriwardena AK, Nadarajah V, de Liguori Carino N. Irreversible electroporation as an intraoperative adjunctive treatment for locally advanced pancreatic cancer after neoadjuvant therapy: An initial clinical experience. Ann Hepatobiliary Pancreat Surg 2025; 29:72-78. [PMID: 39819732 PMCID: PMC11830891 DOI: 10.14701/ahbps.24-193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 01/19/2025] Open
Abstract
Backgrounds/Aims Irreversible electroporation (IRE) may have a potential application as either a "back-up therapy" or for margin accentuation during trial dissection of locally advanced pancreatic cancer (LAPC). The aim of this report was to describe our experience with IRE in terms of its potential applications mentioned above. Methods A clinical policy to use IRE in LAPC patients undergoing exploratory surgery after neoadjuvant therapy (NAT) was initiated in 2017. If resection was feasible, IRE was used for margin accentuation. If not, then IRE was undertaken as a "back-up therapy" of non-resectable tumor. Data on baseline characteristics, perioperative 90-day morbidity, recurrence-free survival (RFS) and overall survival (OS) were collected. Results IRE was successfully performed in 18 (95%) patients. IRE was abandoned in one case for technical reasons. Nine patients who were found to have an unresectable disease underwent IRE as a "back-up therapy" while the remaining patients received IRE for margin accentuation. Complications were recorded in 33% patients. There was no procedure-related mortality. In the group receiving IRE for margin accentuation, the median RFS was 10.0 months (range, 4.5-15.0 months). The median OS of our cohort was 22 months (range, 14.75-27.50 months). Conclusions This report shows that in patients with LAPC undergoing exploratory surgery following NAT, IRE seems technically feasible for margin accentuation or as a "back-up therapy". More data are needed to determine procedure-related morbidity, mortality, and any effects of IRE on cancer-related survival.
Collapse
Affiliation(s)
- Agastya Patel
- Department of Hepato-Pancreato-Biliary Surgery, Manchester Royal Infirmary, Manchester, United Kingdom
- First Doctoral School, Medical University of Gdansk, Gdansk, Poland
| | - Francesco Lancellotti
- Department of Hepato-Pancreato-Biliary Surgery, Manchester Royal Infirmary, Manchester, United Kingdom
| | - Ajith Kumar Siriwardena
- Department of Hepato-Pancreato-Biliary Surgery, Manchester Royal Infirmary, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Vinotha Nadarajah
- Department of Radiology, Manchester Royal Infirmary, Manchester, United Kingdom
| | - Nicola de Liguori Carino
- Department of Hepato-Pancreato-Biliary Surgery, Manchester Royal Infirmary, Manchester, United Kingdom
| |
Collapse
|
23
|
Araki T, Hayashi K, Shimokawa M, Otsuka T, Sonoda Y, Honda T, Shibuki T, Nakazawa J, Arima S, Miwa K, Koga F, Ueda Y, Kubotsu Y, Shimokawa H, Takeshita S, Nishikawa K, Komori A, Otsu S, Hosokawa A, Sakai T, Oda H, Kawahira M, Arita S, Taguchi H, Tsuneyoshi K, Fujita T, Sakae T, Kawaguchi Y, Shirakawa T, Mizuta T, Mitsugi K. Comparison of inflammatory markers before and after nanoliposomal irinotecan and fluorouracil with folic acid in patients with pancreatic cancer: results from the NAPOLEON-2 study (NN-2302). Ther Adv Med Oncol 2025; 17:17588359251320768. [PMID: 39990012 PMCID: PMC11843724 DOI: 10.1177/17588359251320768] [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: 11/21/2024] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
Background Relationships between cancer onset, progression, and inflammation have been reported; yet, the significance of inflammatory markers before and after treatment with nanoliposomal irinotecan and fluorouracil with folic acid (NFF) is unclear. Objectives We aimed to (1) investigate whether worsening inflammation is associated with disease progression and whether it can be used as a biomarker by comparing inflammatory markers before and after treatment with NFF and (2) verify which is a superior biomarker. Design This was a preplanned analysis of a retrospective cohort of the NAPOLEON-2 study, a multicenter observational study. Patients from 20 institutions were enrolled. Those with unresectable pancreatic cancer who received NFF as a second-line or later-line treatment between June 1, 2020, and May 31, 2021, were included. Methods The primary endpoint was overall survival, and the main secondary endpoint was progression-free survival. The following inflammatory markers were assessed: neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio, Prognostic Nutrition Index (PNI), C-reactive protein/albumin ratio (CAR), Glasgow prognostic score (GPS), Prognostic Index (PI), and Komori score. Results A total of 161 patients were enrolled. The CAR, NLR, GPS (1 vs 2), PI (1 vs 2), PNI, and Komori score were useful pre-NFF biomarkers (hazard ratios (HRs): 2.59, 2.28, 2.76/3.35, 2.16/18.89, 2.13, and 3.39, respectively). The CAR, NLR, GPS (compared with a score of 1/2), and PI (compared with a score of 1/2) were useful post-NFF biomarkers (HRs: 2.63, 1.88, 2.54/3.09, 2.30/5.39, respectively). In terms of the receiver operating characteristic curve and Akaike information criterion, the CAR, GPS, and PI showed favorable trends, and overall, CAR was a useful biomarker. Conclusion Inflammatory markers can be used as biomarkers both before and after NFF treatment, and CAR may be useful. We hope these findings will contribute to the selection of appropriate treatment options.
Collapse
Affiliation(s)
- Tomonori Araki
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Nagasaki, Japan
| | - Kohei Hayashi
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Nagasaki, Japan
| | - Mototsugu Shimokawa
- Clinical Research Institute, National Kyushu Cancer Center, Fukuoka-shi, Fukuoka, Japan
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, Ube-shi, Yamaguchi, Japan
| | - Taiga Otsuka
- Department of Internal Medicine, Minato Medical Clinic, 3-11-3 Nagahama, Chuo-ku, Fukuoka-shi, Fukuoka 810-0072, Japan
| | - Yuki Sonoda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Nagasaki, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Nagasaki, Japan
| | - Taro Shibuki
- Department for the Promotion of Drug and Diagnostic Development, Division of Drug and Diagnostic Development Promotion, Translational Research Support Office, National Cancer Center Hospital East, Kashiwa-shi, Chiba, Japan
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa-shi, Chiba, Japan
| | - Junichi Nakazawa
- Department of Medical Oncology, Kagoshima City Hospital, Kagoshima-shi, Kagoshima, Japan
| | - Shiho Arima
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima-shi, Kagoshima, Japan
| | - Keisuke Miwa
- Multidisciplinary Treatment Cancer Center, Kurume University Hospital, Kurume-shi, Fukuoka, Japan
| | - Futa Koga
- Department of Hepatobiliary and Pancreatology, Saga Medical Center Koseikan, Saga-shi, Saga, Japan
| | - Yujiro Ueda
- Department of Hematology and Oncology, Japanese Red Cross Kumamoto Hospital, Kumamoto-shi, Kumamoto, Japan
| | - Yoshihito Kubotsu
- Department of Internal Medicine, Karatsu Red Cross Hospital, Karatsu-shi, Saga, Japan
| | - Hozumi Shimokawa
- Department of Hematology and Oncology, Japan Community Healthcare Organization Kyushu Hospital, Kitakyushu-shi, Fukuoka, Japan
- Department of Medical Oncology, Hamanomachi Hospital, Fukuoka-shi, Fukuoka, Japan
| | - Shigeyuki Takeshita
- Department of Gastroenterology, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki-shi, Nagasaki, Japan
| | - Kazuo Nishikawa
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Yufu-shi, Oita, Japan
| | - Azusa Komori
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Yufu-shi, Oita, Japan
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama-shi, Ehime, Japan
| | - Satoshi Otsu
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Yufu-shi, Oita, Japan
| | - Ayumu Hosokawa
- Department of Clinical Oncology, University of Miyazaki Hospital, Miyazaki-shi, Miyazaki, Japan
| | - Tatsunori Sakai
- Department of Medical Oncology, NHO Kumamoto Medical Center, Kumamoto-shi, Kumamoto, Japan
| | - Hisanobu Oda
- Division of Integrative Medical Oncology, Saiseikai Kumamoto Hospital, Kumamoto-shi, Kumamoto, Japan
| | - Machiko Kawahira
- Department of Gastroenterology, Kagoshima Kouseiren Hospital, Kagoshima-shi, Kagoshima, Japan
| | - Shuji Arita
- Department of Chemotherapy, Miyazaki Prefectural Miyazaki Hospital, Kita-takamatsucho, Miyazaki, Japan
| | - Hiroki Taguchi
- Department of Gastroenterology, Izumi General Medical Center, Izumi-shi, Kagoshima, Japan
- Department of Gastroenterology, Kagoshima City Hospital, Kagoshima-shi, Kagoshima, Japan
| | - Kengo Tsuneyoshi
- Department of Gastroenterology, Izumi General Medical Center, Izumi-shi, Kagoshima, Japan
| | - Toshihiro Fujita
- Department of Gastroenterology, Saiseikai Sendai Hospital, Satsumasendai-shi, Kagoshima, Japan
| | - Takahiro Sakae
- Department of Gastroenterology, Saiseikai Sendai Hospital, Satsumasendai-shi, Kagoshima, Japan
| | - Yasunori Kawaguchi
- Department of Gastroenterology, Asakura Medical Association Hospital, Asakura-shi, Fukuoka, Japan
| | - Tsuyoshi Shirakawa
- Clinical Hematology Oncology Treatment Study Group, Fukuoka-shi, Fukuoka, Japan
- Department of Medical Checkup Center, Eikoh Hospital, Kasuya-gun, Fukuoka, Japan
| | - Toshihiko Mizuta
- Department of Internal Medicine, Fujikawa Hospital, Saga-shi, Saga, Japan
| | - Kenji Mitsugi
- Department of Medical Oncology, Sasebo Kyosai Hospital, Sasebo-shi, Nagasaki, Japan
| |
Collapse
|
24
|
Láinez Ramos-Bossini AJ, Gámez Martínez A, Luengo Gómez D, Valverde-López F, Morillo Gil AJ, González Flores E, Salmerón Ruiz Á, Jiménez Gutiérrez PM, Melguizo C, Prados J. Computed Tomography-Based Sarcopenia and Pancreatic Cancer Survival-A Comprehensive Meta-Analysis Exploring the Influence of Definition Criteria, Prevalence, and Treatment Intention. Cancers (Basel) 2025; 17:607. [PMID: 40002202 PMCID: PMC11853262 DOI: 10.3390/cancers17040607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/16/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Sarcopenia has been associated with poor outcomes in pancreatic cancer (PC). However, published results are heterogeneous in terms of study design, oncological outcomes, and sarcopenia measurements. This meta-analysis aims to evaluate the impact of computed tomography (CT)-based sarcopenia on overall survival (OS) and progression-free survival (PFS) in patients with PC, considering potential confounders such as the CT-based method and thresholds used to define sarcopenia, as well as treatment intention. Methods: We systematically searched databases for observational studies reporting hazard ratios (HRs) for OS and PFS in PC patients stratified by CT-based sarcopenia status. Random-effects models were used to calculate pooled crude and adjusted HRs (cHRs and aHRs, respectively), with subgroup analyses based on sarcopenia measurement methods, cutoff values, sarcopenia prevalence, and treatment intention. Heterogeneity was assessed using the I2 and τ2 statistics, and publication bias was evaluated using funnel plots and Egger's test. Results: Data from 48 studies were included. Sarcopenia was significantly associated with worse OS (pooled cHR = 1.58, 95% CI: 1.38-1.82; pooled aHR = 1.39, 95% CI: 1.16-1.66) and worse PFS (pooled cHR = 1.55, 95% CI: 1.29-1.86; pooled aHR = 1.31, 95% CI: 1.11-1.55). Subgroup analyses revealed significantly different, stronger associations in studies using stricter sarcopenia cutoffs (<50 cm2/m2 for males) and in patients undergoing curative treatments. Heterogeneity was substantial across analyses (I2 > 67%), but with generally low τ2 values (0.01-0.25). Egger's test indicated potential publication bias for OS (p < 0.001), but no significant bias was observed for PFS (p = 0.576). Conclusions: Sarcopenia determined by CT is an independent predictor of poor OS and PFS in PC, but this association varies depending on the cutoff used for its definition as well as on the treatment intention. Therefore, its routine assessment in clinical practice could provide valuable prognostic information, but future research should focus on standardizing sarcopenia assessment methods.
Collapse
Affiliation(s)
- Antonio Jesús Láinez Ramos-Bossini
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
- Advanced Medical Imaging Group (TeCe-22), Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; (C.M.); (J.P.)
| | - Antonio Gámez Martínez
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
| | - David Luengo Gómez
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
- Advanced Medical Imaging Group (TeCe-22), Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
| | - Francisco Valverde-López
- Department of Gastroenterology and Hepatology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain;
| | - Antonio Jesús Morillo Gil
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
| | | | - Ángela Salmerón Ruiz
- Department of Radiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain; (A.G.M.); (D.L.G.); (A.J.M.G.); (Á.S.R.)
- Advanced Medical Imaging Group (TeCe-22), Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
| | | | - Consolación Melguizo
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; (C.M.); (J.P.)
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - José Prados
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; (C.M.); (J.P.)
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| |
Collapse
|
25
|
Aeschbacher P, Wenning AS, Katou S, Morgul H, Juratli M, Becker F, Büdeyri I, Gloor B, Pascher A, Struecker B, Andreou A. Impact of Routine and Selective Preoperative Endoscopic Retrograde Cholangiopancreatography with Stent Placement on Postoperative and Oncologic Outcomes Following Pancreaticoduodenectomy for Pancreatic Ductal Adenocarcinoma. Biomedicines 2025; 13:333. [PMID: 40002746 PMCID: PMC11853506 DOI: 10.3390/biomedicines13020333] [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: 12/27/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Background: According to current guidelines, preoperative endoscopic retrograde cholangiopancreatography (ERCP) with biliary stenting (ERCP/stenting) is often necessary in patients with obstructive jaundice due to pancreatic ductal adenocarcinoma (PDAC), including severe jaundice (bilirubin > 250 umol/l), pruritus, cholangitis, cholestatic liver dysfunction, renal failure, severe malnutrition, or delayed surgery for tumors requiring neoadjuvant chemotherapy. We aimed to investigate the impact of preoperative ERCP/stenting on postoperative and long-term outcomes following pancreaticoduodenectomy (PD) for PDAC. Methods: Clinicopathological data of patients who underwent partial/total PD for PDAC between 2012 and 2019 in two hepato-pancreato-biliary centers in Germany and Switzerland were assessed. We compared patients treated with preoperative ERCP/stenting with those directly undergoing surgery according to postoperative morbidity, postoperative mortality, overall survival (OS) and disease-free survival (DFS). Results: During the study period, 192 patients underwent partial/total PD for PDAC. ERCP/stenting was performed in 105 patients, and 87 patients underwent resection without prior intervention. Postoperative 90-day overall morbidity rate (71% vs. 56%, p = 0.029) and superficial surgical site infection (SSI) rate (39% vs. 17%, p < 0.001) were significantly worse following preoperative ERCP/stenting. Major postoperative morbidity rate (18% vs. 21%, p = 0.650), organ/space SSI rate (7% vs. 14%, p = 0.100), and 90-day postoperative mortality rate (4% vs. 2%, p = 0.549) did not significantly differ between the two groups. After excluding 44 patients for whom the indication for ERCP/stenting was not consistent with current guidelines, ERCP/stenting was associated with a higher superficial SSI rate (36% vs. 17%, p = 0.009) and shorter length of stay (12 vs. 16 days, p = 0.004). Median OS (ERCP/stenting: 18 months vs. no ERCP/stenting: 23 months, p = 0.490) and median DFS (ERCP/stenting: 14 months vs. no ERCP/stenting: 18 months, p = 0.645) were independent from the utilization of ERCP/stenting. Conclusions: Preoperative ERCP/stenting in patients with PDAC can be performed without increasing organ/space SSI, major perioperative morbidity, and mortality rates and without worsening oncologic outcomes. However, it is associated with higher superficial SSI rates. If ERCP/stenting is not performed routinely but according to current guidelines, it is also associated with a shorter length of hospital stay. Further refinement of the indications for preoperative ERCP/stenting may reduce superficial SSI rates.
Collapse
Affiliation(s)
- Pauline Aeschbacher
- Department of Visceral Surgery und Medicine, lnselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Anna Silvia Wenning
- Department of Visceral Surgery und Medicine, lnselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Shadi Katou
- Department of General, Visceral and Transplant Surgery, University Hospital Munster, 48149 Münster, Germany
| | - Haluk Morgul
- Department of General, Visceral and Transplant Surgery, University Hospital Munster, 48149 Münster, Germany
| | - Mazen Juratli
- Department of General, Visceral and Transplant Surgery, University Hospital Munster, 48149 Münster, Germany
| | - Felix Becker
- Department of General, Visceral and Transplant Surgery, University Hospital Munster, 48149 Münster, Germany
| | - Ibrahim Büdeyri
- Department of General, Visceral and Transplant Surgery, University Hospital Munster, 48149 Münster, Germany
| | - Beat Gloor
- Department of Visceral Surgery und Medicine, lnselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Andreas Pascher
- Department of General, Visceral and Transplant Surgery, University Hospital Munster, 48149 Münster, Germany
| | - Benjamin Struecker
- Department of General, Visceral and Transplant Surgery, University Hospital Munster, 48149 Münster, Germany
| | - Andreas Andreou
- Department of General, Visceral and Transplant Surgery, University Hospital Munster, 48149 Münster, Germany
| |
Collapse
|
26
|
Tasia W, Washio A, Yamate K, Morita K, Mori Y, Kahar P, Sasaki R, Ogino C. Catalase-Knockout Complements the Radio-Sensitization Effect of Titanium Peroxide Nanoparticles on Pancreatic Cancer Cells. Molecules 2025; 30:629. [PMID: 39942733 PMCID: PMC11820024 DOI: 10.3390/molecules30030629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/26/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
In previous studies, titanium peroxide nanoparticles (PAA-TiOx NPs) with surfaces functionalized using polyacrylic acid (PAA) and hydrogen peroxide (H2O2) demonstrated a synergistic effect when combined with X-ray irradiation. The combination generated H2O2 and reactive oxygen species (ROS) that enhanced the irradiation efficacy. In the present study, we examined the relationship between catalase and PAA-TiOx NPs sensitization to X-ray radiation because catalase is the primary antioxidant enzyme that converts H2O2 to water and oxygen. Catalase-knockout PANC-1 (dCAT) cells were generated using the CRISPR/Cas9 system, which was confirmed by the suppression of catalase expression in mRNA and protein levels that resulted in an 81.7% decrease in catalase activity compared with levels in wild-type cells. Catalase deficiency was found to increase the production of ROS, particularly in hypoxia. Also, the combination of PAA-TiOx NPs and X-ray 5 Gy resulted in a 7-fold decrease in the survival fraction (SF; p < 0.01) of dCAT cells compared with rates documented in wild-type cells. Interestingly, the combination treatment with X-ray 3 Gy in dCAT cells resulted in an SF similar to that observed in wild-type cells treated with the same combination but at a higher radiation dose (5 Gy). These results suggest that a strategy of catalase inhibition could be used to establish an advanced combination treatment of PAA-TiOx NPs and X-ray irradiation for pancreatic cancer cells.
Collapse
Affiliation(s)
- Winda Tasia
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Kobe 658-8501, Hyogo, Japan; (W.T.); (K.M.); (Y.M.); (P.K.)
- National Research and Innovation Agency (BRIN), Bogor 16911, West Java, Indonesia
| | - Amane Washio
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Kobe 658-8501, Hyogo, Japan; (W.T.); (K.M.); (Y.M.); (P.K.)
| | - Koki Yamate
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Kobe 658-8501, Hyogo, Japan; (W.T.); (K.M.); (Y.M.); (P.K.)
| | - Kenta Morita
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Kobe 658-8501, Hyogo, Japan; (W.T.); (K.M.); (Y.M.); (P.K.)
| | - Yutaro Mori
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Kobe 658-8501, Hyogo, Japan; (W.T.); (K.M.); (Y.M.); (P.K.)
| | - Prihardi Kahar
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Kobe 658-8501, Hyogo, Japan; (W.T.); (K.M.); (Y.M.); (P.K.)
| | - Ryohei Sasaki
- Division of Radiation Oncology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Hyogo, Japan;
| | - Chiaki Ogino
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, Kobe 658-8501, Hyogo, Japan; (W.T.); (K.M.); (Y.M.); (P.K.)
| |
Collapse
|
27
|
Sahu SK, Prabhakar PK, Vyas M. Therapeutical potential of natural products in treatment of pancreatic cancer: a review. Mol Biol Rep 2025; 52:179. [PMID: 39888508 DOI: 10.1007/s11033-025-10287-8] [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: 10/10/2024] [Accepted: 01/22/2025] [Indexed: 02/01/2025]
Abstract
Pancreatic cancer remains as global health challenge, ranking as the seventh leading cause of cancer-related deaths worldwide with high mortality rates and a low five-year survival rate. Despite advancements in conventional therapies, including surgery, chemotherapy, and radiation, the overall survival rates for pancreatic cancer patients have shown minimal improvement. Consequently, there is an urgent need for alternative therapeutic strategies. The search for effective treatments has increasingly turned towards natural products, which offer a diverse array of bioactive compounds with potential anticancer properties. All the natural products, derived from plants, marine organisms, and microorganisms, have emerged as promising candidates in cancer treatment. The review explores the potential role of various natural compounds such as polyphenols, alkaloids, terpenoids, and flavonoids in pancreatic cancer management. With over 60% of cancer medications in clinical trials having natural origins, the review underscores the importance of exploring these compounds for their chemopreventive potential. It covers the epidemiological, molecular pathways influenced by these natural products (such as apoptosis, cell cycle regulation and signaling pathways) and therapeutic aspects aims to contribute to the ongoing efforts in understanding and addressing the complexities of pancreatic cancer. Overall, this review highlights the urgency of developing novel therapeutic strategies and incorporating natural compounds into current treatment modalities to improve outcomes for pancreatic cancer patients.
Collapse
Affiliation(s)
- Sanjeev Kumar Sahu
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| | - Pranav Kumar Prabhakar
- Research and Development Cell, Parul University, P.O. Limda, Dist. Vadodara, Ta.Waghodia, Gujarat, 391760, India
| | - Manish Vyas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| |
Collapse
|
28
|
Chen P, Hou W, Li C, Liang Q, Ma L, Zhao X, Yi C. Lived experiences of patients with advanced pancreatic cancer on patient-reported outcomes (PROs) management: a qualitative phenomenological study in Southwest China. BMJ Open 2025; 15:e084259. [PMID: 39880447 PMCID: PMC11781101 DOI: 10.1136/bmjopen-2024-084259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 01/08/2025] [Indexed: 01/31/2025] Open
Abstract
OBJECTIVES To explore the lived experiences of patients with advanced pancreatic cancer enrolled in a patient-reported outcomes (PROs) management programme and to preliminarily understand how PROs management influences various aspects of patient care and overall quality of life. DESIGN A qualitative phenomenological study. SETTING A national cancer care centre in Southwest China specialised in cancer care, with a comprehensive PROs management programme. PARTICIPANTS 15 participants diagnosed with advanced pancreatic cancer. RESULTS Five key themes emerged from our interviews, including enhanced communication with healthcare providers, attributed to the structured nature of PROs; increased perceived sense of care, with patients feeling more valued and heard; PROs management facilitated better treatment decision-making, with patients feeling more involved and empowered; improved communication with family members, aiding in better understanding and support; and varied perceptions of the impact on quality of life, with some noting improvements in specific aspects like symptom management, while others were uncertain about the overall benefit. CONCLUSIONS PROs management plays a significant role in improving communication between patients with advanced pancreatic cancer and their healthcare providers, enhancing patients' involvement in treatment decisions, and potentially improving family dynamics. However, the impact of PROs management on the overall quality of life of patients remains complex and individualised. The findings suggest that further research with a more diverse patient population is needed to fully understand the implications of PROs management in advanced cancer care.
Collapse
Affiliation(s)
- Ping Chen
- Department of Oncology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan, China
| | - Wanting Hou
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Changlin Li
- Department of Oncology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan, China
| | - Qingyue Liang
- Clinical Nutrition Department, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan, China
| | - Li Ma
- Department of Oncology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan, China
| | - Xiumei Zhao
- Department of Oncology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan, China
| | - Cheng Yi
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
29
|
Andel PCM, van Goor IWJM, Augustinus S, Berrevoet F, Besselink MG, Bhojwani R, Boggi U, Bouwense SAW, Cirkel GA, van Dam JL, Djanani A, Dorcaratto D, Dreyer S, den Dulk M, Frigerio I, Ghorbani P, Goetz MR, Groot Koerkamp B, Gryspeerdt F, Hidalgo Salinas C, Intven M, Izbicki JR, Jorba Martin R, Kauffmann EF, Klug R, Liem MSL, Luyer MDP, Maglione M, Martin-Perez E, Meerdink M, de Meijer VE, Nieuwenhuijs VB, Nikov A, Nunes V, Pando Rau E, Radenkovic D, Roeyen G, Sanchez-Bueno F, Serrablo A, Sparrelid E, Tepetes K, Thakkar RG, Tzimas GN, Verdonk RC, ten Winkel M, Zerbi A, Groot VP, Molenaar IQ, Daamen LA, van Santvoort HC. Routine Imaging or Symptomatic Follow-Up After Resection of Pancreatic Adenocarcinoma. JAMA Surg 2025; 160:74-84. [PMID: 39504033 PMCID: PMC11541741 DOI: 10.1001/jamasurg.2024.5024] [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: 06/26/2024] [Accepted: 08/29/2024] [Indexed: 11/09/2024]
Abstract
IMPORTANCE International guidelines lack consistency in their recommendations regarding routine imaging in the follow-up after pancreatic resection for pancreatic ductal adenocarcinoma (PDAC). Consequently, follow-up strategies differ between centers worldwide. OBJECTIVE To compare clinical outcomes, including recurrence-focused treatment and survival, in patients with PDAC recurrence who received symptomatic follow-up or routine imaging after pancreatic resection in international centers affiliated with the European-African Hepato-Pancreato-Biliary Association (E-AHPBA). DESIGN, SETTING, AND PARTICIPANTS This was a prospective, international, cross-sectional study. Patients from a total of 33 E-AHPBA centers from 13 countries were included between 2020 and 2021. According to the predefined study protocol, patients who underwent PDAC resection and were diagnosed with disease recurrence were prospectively included. Patients were stratified according to postoperative follow-up strategy: symptomatic follow-up (ie, without routine imaging) or routine imaging. EXPOSURES Symptomatic follow-up or routine imaging in patients who underwent PDAC resection. MAIN OUTCOMES AND MEASURES Overall survival (OS) was estimated with Kaplan-Meier curves and compared using the log-rank test. To adjust for potential confounders, multivariable logistic regression was used to evaluate the association between follow-up strategy and recurrence-focused treatment. Multivariable Cox proportional hazard analysis was used to study the independent association between follow-up strategy and OS. RESULTS Overall, 333 patients (mean [SD] age, 65 [11] years; 184 male [55%]) with PDAC recurrence were included. Median (IQR) follow-up at time of analysis 2 years after inclusion of the last patient was 40 (30-58) months. Of the total cohort, 98 patients (29%) received symptomatic follow-up, and 235 patients (71%) received routine imaging. OS was 23 months (95% CI, 19-29 months) vs 28 months (95% CI, 24-30 months) in the groups who received symptomatic follow-up vs routine imaging, respectively (P = .01). Routine imaging was associated with receiving recurrence-focused treatment (adjusted odds ratio, 2.57; 95% CI, 1.22-5.41; P = .01) and prolonged OS (adjusted hazard ratio, 0.75; 95% CI, 0.56-.99; P = .04). CONCLUSION AND RELEVANCE In this international, prospective, cross-sectional study, routine follow-up imaging after pancreatic resection for PDAC was independently associated with receiving recurrence-focused treatment and prolonged OS.
Collapse
Affiliation(s)
- Paul C. M. Andel
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
- St Antonius Hospital Nieuwegein, Nieuwegein, the Netherlands
| | - Iris W. J. M. van Goor
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Simone Augustinus
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Frederik Berrevoet
- Department of General and HPB Surgery and Liver Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Marc G. Besselink
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Rajesh Bhojwani
- Department of Surgery, Santokba Institute of Digestive Surgical Sciences, Santokba Durlabhji Memorial Hospital, Rajasthan, India
| | - Ugo Boggi
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Stefan A. W. Bouwense
- Department of Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Surgery, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Geert A. Cirkel
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jacob L. van Dam
- Department of Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Angela Djanani
- Department of Internal Medicine, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Dimitri Dorcaratto
- Department of Surgery, Hospital Clínico, University of Valencia, Biomedical Research Institute (INCLIVA), Valencia, Spain
| | - Stephan Dreyer
- Department of Academic Surgery, Glasgow Royal Infirmary, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Marcel den Dulk
- Department of Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Surgery, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Isabella Frigerio
- Pancreatic Surgical Unit, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Poya Ghorbani
- Department of Surgery, Karolinska Institutet, Solna, Sweden
| | - Mara R. Goetz
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg, Hamburg, Germany
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Filip Gryspeerdt
- Department of General and HPB Surgery and Liver Transplantation, Ghent University Hospital, Ghent, Belgium
| | | | - Martijn Intven
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jakob R. Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg, Hamburg, Germany
| | - Rosa Jorba Martin
- Department of Surgery, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
| | - Emanuele F. Kauffmann
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Reinhold Klug
- Department of General-, Visceral- and Vascular Surgery, Community Hospital Horn, Horn, Austria
| | - Mike S. L. Liem
- Department of Surgery, Medical Spectrum Twente, Enschede, the Netherlands
| | - Misha D. P. Luyer
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands
| | - Manuel Maglione
- Department of Visceral, Transplant, and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Elena Martin-Perez
- Department of General and Digestive Surgery, University Hospital La Princesa, Madrid, Spain
| | - Mark Meerdink
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Vincent E. de Meijer
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | | | - Andrej Nikov
- Department of Surgery, Military University Hospital Prague, Prague, Czech Republic
| | - Vitor Nunes
- Department of Surgery, Hospital Prof Doutor Fernando Fonseca EPE, Amadora, Portugal
| | - Elizabeth Pando Rau
- Department of Hepato-Pancreatobiliary and Transplant Surgery, Hospital Vall d’Hebron, Barcelona, Spain
- Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Dejan Radenkovic
- Clinic for Digestive Surgery, University Clinical Centra of Serbia, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Geert Roeyen
- Department of HPB, Endocrine and Transplantation Surgery, Antwerp University Hospital, Antwerp, Belgium
| | - Francisco Sanchez-Bueno
- Department of General and Digestive Surgery, Hospital Clínico Universitario “Virgen de la Arrixaca,” Murcia, Spain
| | - Alejandro Serrablo
- Department of General and Digestive Surgery, Miguel Servet University Hospital, Zaragoza, Spain
| | | | | | - Rohan G. Thakkar
- Department of Surgery, Newcastle Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - George N. Tzimas
- Department of HepatoPancreatoBiliary Surgery, Hygeia Hospital, Marousi, Greece
| | - Robert C. Verdonk
- Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St. Antonius Hospital Nieuwegein, Department of Gastroenterology, Utrecht, the Netherlands
| | | | - Alessandro Zerbi
- Department of Pancreatic Surgery, IRCCS Humanitas Hospital, Rozzano, and Humanitas University, Pieve Emanuele, Italy
| | - Vincent P. Groot
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| | - I. Quintus Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| | - Lois A. Daamen
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hjalmar C. van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| |
Collapse
|
30
|
Galouzis N, Khawam M, Alexander EV, Yallourakis MD, Mesropyan L, Luu C, Khreiss MR, Riall TS. Decision regret and satisfaction with shared decision-making in pancreatic surgery. J Gastrointest Surg 2025; 29:101870. [PMID: 39516121 DOI: 10.1016/j.gassur.2024.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/27/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Pancreatic surgery often does not provide long-term survival in patients with cancer or consistently improve symptoms in benign disease. This study aimed to assess decision regret and satisfaction with the decision-making process among patients who underwent pancreatectomy. METHODS This study administered the Brehaut Decision Regret Scale (DRS), 9-Item Shared Decision-Making Questionnaire (SDM-Q-9), and the European Organization for Research and Treatment of Cancer Core Quality of Life Questionnaire (EORTC QLQ-C30) to all patients who underwent elective pancreatectomies from 2021 to 2023. Decision regret was defined as a DRS of >25. In addition, this study evaluated SDM-Q-9 responses in patients with and without regret. RESULTS A total of 143 patients were included in this study, of whom 71 patients (49.6%) completed the distributed surveys. Demographics, pathology, and major complication rates were similar between responders and nonresponders. The indications for surgery were malignancy (67.6%) and benign disease (32.4%). Decision regret after pancreatic surgery was reported in 18.3% of patients. Patients who experienced regret were younger (50.8 ± 18.7 years [younger group] vs 62.0 ± 14.9 years [older group]; P = .03), more likely to have benign disease (39.1% [benign disease] vs 8.3% [malignant disease]; P < .01), underwent a distal pancreatectomy (34.5% [distal pancreatectomy] vs 7.7% [pancreaticoduodenectomy]; P = .02), or experienced a major complication (36.8% [major complication] vs 11.5% [no major complication]; P = .03). Patients with regret had lower global health (57.1 ± 20.1 [patients with regret] vs 76.2 ± 22.2 [patients without regret]; P < .01) and social function scores (61.5 ± 31.5 [patients with regret] vs 77.6 ± 22.0 [patients without regret]; P = .03) on the EORTC QLQ-C30. Patients with regret were less satisfied with the shared decision-making process. CONCLUSION Strong decision regret was reported in 18% of patients who underwent pancreatectomy. Younger age, distal pancreatectomy, benign indications, and major postoperative complications were associated with regret. Data from the SDM-9 highlight areas for potential improvement to help patients make decisions aligned with their goals of care.
Collapse
Affiliation(s)
- Nicholas Galouzis
- Department of Surgery, The University of Arizona, Tucson, AZ, United States
| | - Maria Khawam
- Department of Surgery, The University of Arizona, Tucson, AZ, United States
| | - Evelyn V Alexander
- Department of Surgery, The University of Arizona, Tucson, AZ, United States
| | - Michael D Yallourakis
- Northwest - Gary Division, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lusine Mesropyan
- Department of Surgery, The University of Arizona, Tucson, AZ, United States
| | - Carrie Luu
- Department of Surgery, The University of Arizona, Tucson, AZ, United States
| | - Mohammad R Khreiss
- Department of Surgery, The University of Arizona, Tucson, AZ, United States
| | - Taylor S Riall
- Department of Surgery, The University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
31
|
Lin L, Deng J, Yu J, Bauden M, Andersson R, Shen X, Ansari D, Xue X. Anoikis-related genes linked with patient outcome in pancreatic cancer. Gene 2024; 930:148868. [PMID: 39154969 DOI: 10.1016/j.gene.2024.148868] [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: 02/06/2024] [Revised: 08/01/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024]
Abstract
Anoikis is programmed cell death occurring upon cell detachment from the extracellular matrix. Cancer cells need to evade anoikis to be able to metastasize to distant sites. However, the molecular features and prognostic value of anoikis-related genes (ARGs) in pancreatic cancer remain unclear. In this study, we utilized transcriptome data from the TCGA and GSE102238 databases to identify 64 ARGs significantly associated with prognosis. We used the "ConsensusClusterPlus" R package to stratify patients into high and low-risk prognostic subgroups. The KEGG and GSEA analyses revealed that the clusters with poor prognosis were enriched for the ECM receptor interaction pathway, the TP53 signaling pathway, and the galactose metabolism pathway, and that the cell cycle pathway was upregulated. A prognostic model consisting of seven ARGs (SERPINE1, EGF, E2F1, MSLN, RAB27B, ETV7, MST1) was constructed using LASSO regression and when combined with clinicopathological parameters using Cox regression, a prognostic Nomogram was created, which demonstrated high prognostic utility. Among the biomarker candidates, we report ETV7 as a novel, independent prognostic marker in pancreatic cancer. ETV7 was highly expressed in KRAS and TP53 co-occurrent mutant TCGA patients, indicating that it may be regulated by the two major driver genes of pancreatic cancer. Therefore, targeting ETV7 could be a potential focus for future therapeutic studies.
Collapse
Affiliation(s)
- Lizhi Lin
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden; Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jing Deng
- Department of Basic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jiaye Yu
- Department of Basic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Monika Bauden
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| | - Roland Andersson
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| | - Xian Shen
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Daniel Ansari
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden.
| | - Xiangyang Xue
- Department of Basic Medicine, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
32
|
Hansen N, Peña-Martínez P, Skoog P, Reinbach K, Hansen FC, Faria SL, Grönberg C, Abdilleh K, Magnusson S, von Wachenfeldt K, Millrud CR, Liberg D, Järås M. Blocking IL1RAP on cancer-associated fibroblasts in pancreatic ductal adenocarcinoma suppresses IL-1-induced neutrophil recruitment. J Immunother Cancer 2024; 12:e009523. [PMID: 39694705 DOI: 10.1136/jitc-2024-009523] [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: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) represents a major clinical challenge due to its tumor microenvironment, which exhibits immune-suppressive properties that facilitate cancer progression, metastasis, and therapy resistance. Interleukin 1 (IL-1) signaling has been implicated as a driver in this process. Mechanistically, both IL-1α and IL-1β bind to the IL-1 receptor type 1, forming a complex with IL-1-receptor accessory protein (IL1RAP), which triggers downstream signaling pathways. The IL1RAP blocking antibody nadunolimab is currently in clinical development, but the precise consequences of inhibiting IL-1 signaling in PDAC remains elusive. METHODS To evaluate the biological relevance of blocking IL1RAP using nadunolimab in a PDAC animal model, human PDAC cells and cancer-associated fibroblasts (CAFs) were co-transplanted into mice. To study the underlying mechanisms of IL1RAP blockade ex vivo, co-cultured PDAC cells and CAFs were treated with nadunolimab prior to RNA sequencing. Migration assays were performed to assess how nadunolimab affects interactions between CAFs and myeloid immune cells. Finally, to establish a clinical correlation between IL1RAP expression and nadunolimab treatment effects, we analyzed tumor biopsies from a clinical phase I/II study in which nadunolimab was administered to patients. RESULTS In the xenograft mouse model, nadunolimab exhibited antitumor effects only when human CAFs were co-transplanted with PDAC cells. IL-1 stimulation induced CAFs to secrete chemokines that recruited neutrophils and monocytes. The secretion of this chemokine and the migration of myeloid cells were inhibited by nadunolimab. Media conditioned by IL-1-stimulated CAFs sustained a neutrophil population with a tissue invasion phenotype, an effect that was reversed by nadunolimab. In a cohort of metastatic late-stage PDAC patients receiving nadunolimab as monotherapy, high IL1RAP expression in tumors was associated with extended progression-free survival. CONCLUSIONS Our study demonstrates that targeting IL1RAP on CAFs inhibits IL-1-induced chemokine secretion and recruitment of neutrophils and monocytes, thereby counteracting the immunosuppressive microenvironment in PDAC. These findings highlight the therapeutic potential of targeting IL1RAP in PDAC.
Collapse
Affiliation(s)
- Nils Hansen
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | | | | | | | | | | | | | - Kawther Abdilleh
- Pancreatic Cancer Action Network, Manhattan Beach, California, USA
| | | | | | | | | | - Marcus Järås
- Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
33
|
Liu C, Wu H, Cai D, Yu X. Unraveling the causal relationship between serum minerals and pancreatic cancer: a Mendelian randomization study. Discov Oncol 2024; 15:788. [PMID: 39692823 DOI: 10.1007/s12672-024-01695-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/11/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Pancreatic cancer is among the most lethal malignancies, characterized by a poor prognosis and limited modifiable factors. Emerging evidence indicates that serum mineral levels may influence the likelihood of developing pancreatic cancer. However, the causal relationship between serum minerals and pancreatic cancer remains unclear and warrants further investigation. METHODS This Mendelian randomization (MR) study was conducted to explore the causal effects of serum mineral levels on pancreatic cancer risk. Genetic variants associated with serum mineral levels, including calcium, iron, magnesium, zinc, selenium, and copper, were selected as instrumental variables (IVs) from large-scale genome-wide association study (GWAS) data. Multiple methods, including inverse variance weighting (IVW), MR-Egger, weighted median, weight methods, were employed to perform MR analysis. The effect sizes from the MR analysis, using two independent GWAS summary datasets related to pancreatic cancer, were combined through meta-analysis. The Cochrane Q test, MR-Egger intercept test, MR-PRESSO global test, and leave-one-out test were conducted for sensitivity tests. RESULTS Our MR analysis demonstrated a significant causal effect of genetically predicted serum calcium levels on increased pancreatic cancer risk [OR = 1.64, 95% CI 1.05-2.57, P = 0.029 (discovery cohort); OR = 1.52, 95% CI 1.07-2.15, P = 0.019 (validation cohort)], while no significant associations were found for other serum minerals (P > 0.05). Additional meta-analysis reinforces and substantiates this conclusion (pooled OR = 1.56, 95% CI 1.19-2.06, P = 0.001). No evidence of pleiotropy or heterogeneity was detected across multiple sensitivity tests (P > 0.05). CONCLUSION This study provides new evidence supporting the causal role of certain serum minerals, particularly calcium, in the development of pancreatic cancer. These findings may help inform future research into preventive strategies or therapies aimed at modulating mineral levels in patients at high risk of pancreatic cancer.
Collapse
Affiliation(s)
- Cong Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Huajun Wu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Dongdong Cai
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xin Yu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
34
|
Nakamura Y, Yasukawa T, Fukumura Y, Takeda Y, Imamura H, Shi Y, Li M, Abe M, Uyama S, Kajino K, Ishijima M, Saiura A, Orimo A. Association of stromal type IV collagen and prognosis in neoadjuvant chemotherapy-treated pancreatic cancer. Jpn J Clin Oncol 2024; 54:1261-1271. [PMID: 39180719 PMCID: PMC11631120 DOI: 10.1093/jjco/hyae118] [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: 05/31/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has poor prognosis due to its low surgical eligibility and resistance to chemotherapy. Abundant stroma is characteristic of PDAC, and cancer-associated fibroblasts (CAFs) are a major stromal constituent, contributing to chemoresistance. Because neoadjuvant chemotherapy (NAC) is included in PDAC treatment as a standard regimen, the role of CAFs in NAC resistance must be studied. Although type IV collagen (COLIV) is present in the tumor of PDAC, the association between COLIV and disease advancement of NAC-treated PDAC is unclear. METHODS Using a cohort of NAC-treated patients with PDAC, we examined clinicopathological data and conducted immunohistochemical analysis of COLIV in tissue specimens prepared from surgically resected pancreas. RESULTS AND CONCLUSIONS Our analysis revealed that ~50% of the cases were positive for COLIV in the stroma and diffuse COLIV staining was an independent poor prognosis factor alongside high serum CA19-9 before NAC treatment (>37 U/mL) and postsurgical residual tumors. Based on these findings, we propose that stromal COLIV staining can be used to predict prognosis in NAC-treated patients with PDAC after surgery. Additionally, these findings suggest a possibility that stromal COLIV staining indicates resistance to anticancer drugs and/or contributes to malignancy in PDAC.
Collapse
Affiliation(s)
- Yasuhiro Nakamura
- Department of Orthopaedics, Faculty of Medicine, Juntendo University, Tokyo, Japan
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Molecular Pathogenesis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takehiro Yasukawa
- Department of Molecular Pathogenesis, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Pathology and Oncology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yuki Fukumura
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoshinori Takeda
- Department of Hepatobiliary and Pancreatic Surgery, Juntendo University School of Medicine, Tokyo Japan
| | - Hiroshi Imamura
- Department of Hepatobiliary and Pancreatic Surgery, Juntendo University School of Medicine, Tokyo Japan
| | - Yang Shi
- Department of Molecular Pathogenesis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mu Li
- Department of Molecular Pathogenesis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masaaki Abe
- Department of Pathology and Oncology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Saya Uyama
- Department of Pathology and Oncology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Kazunori Kajino
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Muneaki Ishijima
- Department of Orthopaedics, Faculty of Medicine, Juntendo University, Tokyo, Japan
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akio Saiura
- Department of Hepatobiliary and Pancreatic Surgery, Juntendo University School of Medicine, Tokyo Japan
| | - Akira Orimo
- Department of Molecular Pathogenesis, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Pathology and Oncology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
35
|
Wang D, Cao J, Chen Y, Zhang L, Zhou C, Huang L, Chen Y. Radioresistance-related gene signatures identified by transcriptomics characterize the prognosis and immune landscape of pancreatic cancer patients. BMC Cancer 2024; 24:1497. [PMID: 39639217 PMCID: PMC11619475 DOI: 10.1186/s12885-024-13231-4] [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: 07/31/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Radiotherapy (RT) is an important means of local treatment of solid tumors, and radioresistance is the main reason for RT failure for tumors, especially pancreatic cancer (PC). It is urgent to distinguish key genes and mechanisms of radioresistance in PC. METHODS We acquired the data from The Cancer Genome Atlas (TCGA), obtained the gene modules associated with radioresistance by weighted gene coexpression network analysis (WGCNA), and identified differentially expressed genes (DEGs) between normal and tumor samples. Radioresistance-related genes (RRRGs) were determined with the intersection of WGCNA and DEGs. The hub RRRGs associated with prognosis were distinguished by the least absolute shrinkage and selection operator (LASSO) regression. We established a risk score model using multivariate Cox regression. Immune cell infiltration and drug sensitivity were evaluated through the CIBERSORT algorithm and the "OncoPredict" software package, respectively. The association of the key gene RIC3 and PC clinical features was verified in public databases, and its biological behaviors were explored in vitro. RESULTS The intersection of DEGs and WGCNA confirmed 14 RRRGs, then six hub RRRGs were identified using LASSO. A key four genes (DUSP4, ADORA2B, SCGB2A1, and RIC3) risk score model was constructed and proved to be capable of independently estimating the prognosis of PC. There is no significant difference between risk score groups in various immune cell infiltration and response to immunotherapy. Although the low-risk group seemed to exhibit greater sensitivity to antitumor drugs, the four drugs (5-fluorouracil [5-FU], leucovorin, irinotecan, and oxaliplatin) currently used for PC patients had no statistical difference for the low- and high- group. The overexpression of RIC3 had a synergy effect with irradiation on inhibited malignant biological properties of PC cells, which was verified by detecting the proliferation ability, apoptosis rate, cell cycle distribution, and migration ability of PANC-1 cells. CONCLUSIONS We herein presented signature genes correlated with radioresistance in PC and established a risk score model competent in estimating patients' clinical outcomes and response to antitumor drugs. The above evidence could contribute to comprehending the mechanisms of radioresistance and identifying the underlying therapy targeting.
Collapse
Affiliation(s)
- Dandan Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Jun Cao
- Department of Outpatient, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yanhui Chen
- Department of Neuroscience and Endocrinology, Tangshan Caofeidian District Hospital, Tangshan, Hebei, China
| | - Lisha Zhang
- Department of Obstetrics, Tangshan Caofeidian District Hospital, Tangshan, Hebei, China
| | - Chan Zhou
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Litao Huang
- Department of Clinical Research Management, West China Hospital of Sichuan University, Chengdu, China
| | - Yanliang Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China.
- The First School of Clinical Medicine, Lanzhou University, No. 1, Donggang West Road, Chengguan District, Lanzhou, 730000, China.
| |
Collapse
|
36
|
Lin HT, Krishna SG. Understanding the risk of recurrence after resection of intraductal papillary mucinous neoplasm-associated adenocarcinoma: insights from a large multicenter study. Hepatobiliary Surg Nutr 2024; 13:1041-1044. [PMID: 39669079 PMCID: PMC11634424 DOI: 10.21037/hbsn-24-583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 10/30/2024] [Indexed: 12/14/2024]
Affiliation(s)
- Hong T. Lin
- Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Somashekar G. Krishna
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
37
|
Saha B, Chakravarty S, Ray S, Saha H, Das K, Ghosh I, Mallick B, Biswas NK, Goswami S. Correlating tissue and plasma‑specific piRNA changes to predict their possible role in pancreatic malignancy and chronic inflammation. Biomed Rep 2024; 21:186. [PMID: 39420923 PMCID: PMC11484194 DOI: 10.3892/br.2024.1874] [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: 07/01/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
The aggressiveness of pancreatic ductal adenocarcinoma is primarily due to lack of effective early detection biomarkers. Circulating non-coding RNAs serve as diagnostic or prognostic biomarkers in multiple types of cancer. Comparison of their expression between diseased tissue and relevant body fluids such as saliva, urine, bile, pancreatic juice, blood etc. may reveal mechanistic involvement of common non-coding RNAs. piwi-interacting RNAs (piRNAs) are a class of non-coding RNAs. The aim of the present study was to investigate plasma and tumour tissue piRNA changes in patients with pancreatic cancer (PC) and explore the possible role in tumorigenesis and pancreatic inflammation. Sequencing of circulating plasma small RNAs from patients with PC and chronic pancreatitis (CP) was performed and differentially expressed piRNAs were compared with those in tissues. Subsequent search for target genes for those piRNAs was performed followed by pathway and cluster analysis. A total of 36 piRNAs were shown to be deregulated in pancreatic tumour tissue and alteration of 11 piRNAs was detected in plasma of patients with PC. piRNAs hsa-piR-23246, hsa-piR-32858 and hsa-piR-9137 may serve a key role in PC development as their expression was correlated in both plasma and tumour tissue. Key piRNA-target interactions interfering with key biological pathways were also characterized. A total of 19 deregulated piRNAs in plasma samples of patients with CP was identified; these targeted genes responsible for chronic inflammation. Therefore, the present study provides a comprehensive description of piRNA alteration in pancreatic malignancy and inflammation; these may be explored for biomarker potential in future.
Collapse
Affiliation(s)
- Barsha Saha
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, Nadia, West Bengal 741251, India
- Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad 121001, India
| | - Shouvik Chakravarty
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, Nadia, West Bengal 741251, India
- Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad 121001, India
| | - Sukanta Ray
- Institute of Postgraduate Medical Education & Research, Kolkata 700020, India
| | - Hemabha Saha
- Institute of Postgraduate Medical Education & Research, Kolkata 700020, India
| | - Kshaunish Das
- Institute of Postgraduate Medical Education & Research, Kolkata 700020, India
| | - Indranil Ghosh
- Chittaranjan National Cancer Institute, Kolkata 700026, India
| | | | - Nidhan K. Biswas
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, Nadia, West Bengal 741251, India
| | - Srikanta Goswami
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, Nadia, West Bengal 741251, India
| |
Collapse
|
38
|
Kang YK, Ha S, Jeong JB, Oh SW. The value of PET/CT radiomics for predicting survival outcomes in patients with pancreatic ductal adenocarcinoma. Sci Rep 2024; 14:28958. [PMID: 39578496 PMCID: PMC11584753 DOI: 10.1038/s41598-024-77022-4] [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: 07/30/2024] [Accepted: 10/18/2024] [Indexed: 11/24/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with poor prognosis even without distant metastases, necessitating in-depth characterization of primary tumors for survival prediction. We assessed the feasibility of using FDG-PET/CT radiomics to predict overall survival (OS) in PDAC. This retrospective study included PDAC patients without distant metastasis who underwent FDG-PET/CT for initial staging. Primary tumors were segmented from FDG-PET/CT images, extracting 222 radiomics features. A radiomics-based risk score (Rad-score) was developed using Cox proportional hazards regression with LASSO to predict OS. The prognostic performance of the Rad-score was compared with a clinical model (demographics, disease stage, laboratory results) using Harrell's concordance index (C-index) and bootstrapping. 140 patients were included, with a mortality rate was 72.9% during follow-up (total population, 19.5 ± 19.2 months; survivors, 34.4 ± 28.8 months). Eleven radiomics features were significant for survival prediction. The Rad-score predicted OS with a C-index of 0.681 [95% CI, 0.632-0.731]. A model integrating clinical parameters and Rad-score outperformed the clinical-only model in predicting OS (C-index 0.740 [0.715-0.816] vs. 0.673 [0.650-0.766]; C-index difference 0.067 [0.014-0.113]; P < 0.001). These findings suggest that incorporating FDG-PET/CT radiomics into preexisting prognotic stratification paradiagm may enhance survival prediction in PDAC, warranting large-scale studies to confirm its applicability in clinical practice.
Collapse
Affiliation(s)
- Yeon-Koo Kang
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Republic of Korea
| | - Ji Bong Jeong
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul Metropolitan Government - Seoul National University Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-Gu, Seoul, Republic of Korea.
| | - So Won Oh
- Department of Nuclear Medicine, Seoul Metropolitan Government - Seoul National University Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-Gu, Seoul, Republic of Korea.
| |
Collapse
|
39
|
Cho YS, Cho H, Kim HR, Park SJ, Yeo JH, Ko YG, Lee J, Kim SY, Kim K, Byun Y. Macropinocytosis-targeted peptide-docetaxel conjugate for bystander pancreatic cancer treatment. J Control Release 2024; 376:829-841. [PMID: 39491626 DOI: 10.1016/j.jconrel.2024.10.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Oncogenic Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations are highly prevalent in pancreatic ductal adenocarcinoma (PDAC) and have garnered attention as potential targets for targeted therapies, such as KRAS inhibitors. However, the limited therapeutic efficacy of KRAS allele-specific inhibitors necessitate an efficient pan-KRAS cancer cell killing strategy. Here, we have examined enhanced macropinocytosis pathway in KRAS mutant cancer cells and report improved intracellular delivery of albumin-based therapeutics. We further established an albumin-binding peptide-docetaxel conjugate platform (MPD3), which has a caspase-3 cleavable feature, for macropinocytosis-targeted bystander payload delivery and realization of bystander killing of pan-KRAS cancer cells, complemented with caspase-3 mediated activation of MPD3 to bolster tumoral accumulation of cytotoxic payloads. Utilization of in vitro co-culture system of pan-KRAS cancer cells and pharmacodynamic marker staining revealed potent bystander killing effects of MPD3, highlighting MPD3 as an efficient delivery platform against pan-KRAS cancer. Moreover, MPD3 elicited robust anti-tumor activities in both local and liver metastatic PDAC tumor models in mice. Overall, this work establishes a paradigm for developing translational pan-KRAS cancer treatment and broadens the applicability of albumin binding peptide-drug conjugate against albumin-metabolism enriched cancers.
Collapse
Affiliation(s)
- Young Seok Cho
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergent Science and Technology, Seoul National University, Seoul 08826, Republic of Korea; Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hanhee Cho
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Ha Rin Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; School of Medicine, Stanford University, CA 94305, USA
| | - Seong Jin Park
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Joo Hye Yeo
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, Republic of Korea
| | - Yoon Gun Ko
- Pharosgen Co.Ltd, 2-404 Jangji-dong 892, Seoul 05852, Republic of Korea
| | - Jinu Lee
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, Republic of Korea
| | - Sang Yoon Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Kwangmeyung Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Youngro Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
40
|
Qadir RMAB, Umair MB, Tariq UB, Ahmad A, Kiran W, Shahid MH. Unraveling Pancreatic Cancer: Epidemiology, Risk Factors, and Global Trends. Cureus 2024; 16:e72816. [PMID: 39493341 PMCID: PMC11528318 DOI: 10.7759/cureus.72816] [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] [Accepted: 11/01/2024] [Indexed: 11/05/2024] Open
Abstract
Pancreatic cancer is one of the most lethal malignancies, characterized by late diagnosis, rapid progression, and limited treatment options. This literature review comprehensively examines the epidemiology, risk factors, diagnostic challenges, treatment modalities, and prognosis of pancreatic cancer. It highlights the global disparities in incidence and outcomes, exploring the influence of socioeconomic, environmental, and genetic factors on disease progression. In addition, this review discusses recent advancements in diagnostic tools and treatment strategies, identifying gaps in current research and clinical practices. The synthesis aims to inform future research directions and policy-making efforts to reduce the global burden of pancreatic cancer and improve patient outcomes.
Collapse
Affiliation(s)
| | | | - Umar Bin Tariq
- General Surgery, Southmead Hospital Bristol, North Bristol NHS Trust, Bristol, GBR
| | - Arslan Ahmad
- Emergency Medicine, Weston General Hospital, University Hospitals Bristol and Weston NHS Foundation Trust, Weston-super-Mare, GBR
| | - Wajeeha Kiran
- Trauma and Orthopaedics, Morriston Hospital, Swansea, GBR
| | | |
Collapse
|
41
|
Pratt EC, Mezzadra R, Kulick A, Kaminsky S, Samuels ZV, Loor A, de Stanchina E, Lowe SW, Lewis JS. uPAR Immuno-PET in Pancreatic Cancer, Aging, and Chemotherapy-Induced Senescence. J Nucl Med 2024; 65:1718-1723. [PMID: 39362768 PMCID: PMC11533913 DOI: 10.2967/jnumed.124.268278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/13/2024] [Indexed: 10/05/2024] Open
Abstract
Identifying cancer therapy resistance is a key time-saving tool for physicians. Part of chemotherapy resistance includes senescence, a persistent state without cell division or cell death. Chemically inducing senescence with the combination of trametinib and palbociclib (TP) yields several tumorigenic and prometastatic factors in pancreatic cancer models with many potential antibody-based targets. In particular, urokinase plasminogen activator receptor (uPAR) has been shown to be a membrane-bound marker of senescence in addition to an oncology target. Methods: Here, 2 antibodies against murine uPAR and human uPAR were developed as immuno-PET agents to noninvasively track uPAR antigen abundance. Results: TP treatment increased cell uptake both in murine KPC cells and in human MiaPaCa2 cells. In vivo, subcutaneously implanted murine KPC tumors had high tumor uptake with the antimurine uPAR antibody independently of TP in young mice, yet uPAR uptake was maintained in aged mice on TP. Mice xenografted with human MiaPaCa2 tumors showed a significant increase in tumor uptake on TP therapy when imaged with the antihuman uPAR antibody. Imaging with either uPAR antibody was found to be more tumor-selective than imaging with [18F]FDG or [18F]F-DPA-714. Conclusion: The use of radiolabeled uPAR-targeting antibodies provides a new antibody-based PET imaging candidate for pancreatic cancer imaging as well as chemotherapy-induced senescence.
Collapse
Affiliation(s)
- Edwin C Pratt
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Riccardo Mezzadra
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amanda Kulick
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Spencer Kaminsky
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zachary V Samuels
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Angelique Loor
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- HHMI, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York;
- Department of Pharmacology, Weill Cornell Graduate School, New York, New York
| |
Collapse
|
42
|
van Eijck CWF, Ju J, van 't Land FR, Verheij M, Li Y, Stubbs A, Doukas M, Lila K, Heij LR, Wiltberger G, Alonso L, Malats N, Groot Koerkamp B, Vietsch EE, van Eijck CHJ. The tumor immune microenvironment in resected treatment-naive pancreatic cancer patients with long-term survival. Pancreatology 2024; 24:1057-1065. [PMID: 39218754 DOI: 10.1016/j.pan.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/27/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers worldwide. Presently, only a fraction of patients undergo successful surgical resection, the most effective treatment. Enhancing treatment strategies necessitates a deep comprehension of the factors underlying extended survival after surgical resection in patients. METHODS This study aims to identify the important factors of PDAC patients' long-term survival with metatranscriptomics and multiplex immunofluorescence (IF) staining analyses. Specifically, differences in tumor immune microenvironment (TIME) were investigated between treatment-naïve PDAC short-term survivors (STS, overall survival <6 months) and long-term survivors (LTS, overall survival >5 years). RESULTS As a result, we detected 589 over-expressed genes, including HOXB9, CDA, and HOXB8, and 507 under-expressed genes, including AMY2B, SCARA5, and SLC2A2 in LTS. Most of the Reactome overbiological pathways enriched in our data were over-expressed in LTS, such as RHO GTPase Effectors and Cell Cycle Checkpoints. Eleven microbiomes significantly differed between LTS and STS, including Sphingopyxis and Capnocytophaga. Importantly, we demonstrate that the TIME profile with an increased abundance of memory B cells and the reduction of M0 and pro-tumoral M2 macrophages are associated with a good prognosis in PDAC. CONCLUSIONS In this study, we delved into the TIME with metatranscriptomics and IF staining analyses to understand the prerequisite of prolonged survival in PDAC patients. In LTS, several biological pathways were overexpressed, and specific microbiomes were identified. Furthermore, apparent differences in driven immune factors were found that provide valuable insights into developing new treatment strategies.
Collapse
Affiliation(s)
- Casper W F van Eijck
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
| | - Jie Ju
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Freek R van 't Land
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Maaike Verheij
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Yunlei Li
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Andrew Stubbs
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Michael Doukas
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Karishma Lila
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Lara R Heij
- Institute of Pathology, Medical Center University Duisburg-Essen, Essen, Germany; Department of Surgery and Transplantation, University Hospital Essen, Germany; Department of General, Gastrointestinal, Hepatobiliary and Transplant Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Georg Wiltberger
- Department of General, Gastrointestinal, Hepatobiliary and Transplant Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Lola Alonso
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Bas Groot Koerkamp
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Eveline E Vietsch
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Casper H J van Eijck
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
43
|
Zheng Y, Yang Y, Xiong Q, Ma Y, Zhu Q. Establishment and Verification of a Novel Gene Signature Connecting Hypoxia and Lactylation for Predicting Prognosis and Immunotherapy of Pancreatic Ductal Adenocarcinoma Patients by Integrating Multi-Machine Learning and Single-Cell Analysis. Int J Mol Sci 2024; 25:11143. [PMID: 39456925 PMCID: PMC11508839 DOI: 10.3390/ijms252011143] [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: 08/25/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has earned a notorious reputation as one of the most formidable and deadliest malignant tumors. Within the tumor microenvironment, cancer cells have acquired the capability to maintain incessant expansion and increased proliferation in response to hypoxia via metabolic reconfiguration, leading to elevated levels of lactate within the tumor surroundings. However, there have been limited studies specifically investigating the association between hypoxia and lactic acid metabolism-related lactylation in PDAC. In this study, multiple machine learning approaches, including LASSO regression analysis, XGBoost, and Random Forest, were employed to identify hub genes and construct a prognostic risk signature. The implementation of the CERES score and single-cell analysis was used to discern a prospective therapeutic target for the management of PDAC. CCK8 assay, colony formation assays, transwell, and wound-healing assays were used to explore both the proliferation and migration of PDAC cells affected by CENPA. In conclusion, we discovered two distinct subtypes characterized by their unique hypoxia and lactylation profiles and developed a risk score to evaluate prognosis, as well as response to immunotherapy and chemotherapy, in PDAC patients. Furthermore, we indicated that CENPA may serve as a promising therapeutic target for PDAC.
Collapse
Affiliation(s)
| | | | | | | | - Qing Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; (Y.Z.)
| |
Collapse
|
44
|
Wu D, Wang Z, Zhang Y, Yang Y, Yang Y, Zu G, Yu X, Chen W, Qin Y, Xu X, Chen X. IL15RA-STAT3-GPX4/ACSL3 signaling leads to ferroptosis resistance in pancreatic cancer. Acta Biochim Biophys Sin (Shanghai) 2024; 57:389-402. [PMID: 39396119 PMCID: PMC11986442 DOI: 10.3724/abbs.2024153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/05/2024] [Indexed: 10/14/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant disease with a poor prognosis, and the lack of effective treatment methods accounts for its high mortality. Pancreatic stellate cells (PSCs) in the tumor microenvironment play an important role in the development of PDAC. Previous studies have reported that patients with PDAC are more vulnerable to ferroptosis inducers. To investigate the relationship between PSCs and pancreatic cancer cells, a coculture system is used to further reveal the influence of PSCs on ferroptosis resistance in PDAC using many in vitro and in vivo experiments. Our results show that PSCs promote ferroptosis resistance in pancreatic cancer cells. We further demonstrate that IL15 secretion by PSCs activates the IL15RA-STAT3-GPX4/ACSL3 axis. The simultaneous upregulation of GPX4 and ACSL3 prevents lipid peroxidation and ultimately protects pancreatic cancer cells from ferroptosis both in vitro and in vivo. This study demonstrates that PSCs protect pancreatic cancer cells in a paracrine manner and may indicate a novel strategy for the treatment of PDAC.
Collapse
Affiliation(s)
- Di Wu
- Department of Hepatopancreatobiliarythe Third Affiliated Hospital of Soochow UniversityChangzhou213000China
| | - Zhiliang Wang
- Department of Hepatopancreatobiliarythe Third Affiliated Hospital of Soochow UniversityChangzhou213000China
| | - Yue Zhang
- Department of Hepatopancreatobiliarythe Third Affiliated Hospital of Soochow UniversityChangzhou213000China
| | - Yang Yang
- Department of Hepatopancreatobiliarythe Third Affiliated Hospital of Soochow UniversityChangzhou213000China
| | - Yue Yang
- Department of Hepatopancreatobiliarythe Third Affiliated Hospital of Soochow UniversityChangzhou213000China
| | - Guangchen Zu
- Department of Hepatopancreatobiliarythe Third Affiliated Hospital of Soochow UniversityChangzhou213000China
| | - Xianjun Yu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Shanghai Pancreatic Cancer InstituteShanghai200032China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032China
| | - Weibo Chen
- Department of Hepatopancreatobiliarythe Third Affiliated Hospital of Soochow UniversityChangzhou213000China
| | - Yi Qin
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Shanghai Pancreatic Cancer InstituteShanghai200032China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032China
| | - Xiaowu Xu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Shanghai Pancreatic Cancer InstituteShanghai200032China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032China
| | - Xuemin Chen
- Department of Hepatopancreatobiliarythe Third Affiliated Hospital of Soochow UniversityChangzhou213000China
| |
Collapse
|
45
|
Yang HY, Shun SC, Lee YH, Liou YT, Chou YJ, Kuo HJ, Tien YW, Lai SR, Hung H. Trajectories of perioperative nutritional status in patients with pancreatic tumor after surgery in six months. Eur J Oncol Nurs 2024; 72:102687. [PMID: 39288676 DOI: 10.1016/j.ejon.2024.102687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/18/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE To characterize subgroups with similar nutritional status trajectories during the 6-month period after pancreatectomy and to identify demographic and clinical characteristics influencing changes in nutritional status in each subgroup. METHODS This longitudinal prospective study recruited 112 patients with newly diagnosed pancreatic tumor from an outpatient pancreatic surgical department of a medical center in northern Taiwan between September 2016 and April 2019. Patients completed a demographic and clinical characteristics form, the Mini Nutritional Assessment scale, and the Symptom Severity Scale prior to surgery (T0), 3 months after surgery (T1), and 6 months after surgery (T2). Latent class growth analysis was used to investigate the trajectories of nutritional status. Generalized estimating equations were used to identify significant factors influencing each trajectory. RESULTS Two latent groups of nutritional status trajectories were identified. Among 112 patients, 74.11% and 25.89% were classified as having high and low nutritional status trajectories, respectively. High nutritional status was significantly negatively correlated with changes in symptom severity. Low nutritional status was significantly negatively correlated with older age, surgical complications, and changes in symptom severity. CONCLUSIONS Symptom severity has the most significant negative effect on perioperative nutritional status. Older age and surgical complications exert negative effects on perioperative nutritional status among patients with low nutritional status. These findings emphasize the need for nurses to identify at-risk individuals and provide individualized nutritional care to improve nutritional status in this population. CLINICAL TRIALS REGISTRATION This study was registered on ClinicalTrials.gov (trial registration number: NCT02900677; approved date: September 14th, 2016). Link: https://clinicaltrials.gov/ct2/show/NCT02900677.
Collapse
Affiliation(s)
- Hui-Ying Yang
- Institute of Clinical Nursing, College of Nursing, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shiow-Ching Shun
- Institute of Clinical Nursing, College of Nursing, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Yun-Hsiang Lee
- School of Nursing, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yan-Ting Liou
- National Taiwan University Cancer Center, Taipei, Taiwan
| | - Yun-Jen Chou
- School of Nursing, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsuan-Ju Kuo
- School of Nursing, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Wen Tien
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Sheng-Ru Lai
- Department of Dietetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Hung Hung
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
46
|
Merali N, Chouari T, Sweeney C, Halle-Smith J, Jessel MD, Wang B, O’ Brien J, Suyama S, Jiménez JI, Roberts KJ, Velliou E, Sivakumar S, Rockall TA, Demirkan A, Pedicord V, Deng D, Giovannetti E, Annels NE, Frampton AE. The microbial composition of pancreatic ductal adenocarcinoma: a systematic review of 16S rRNA gene sequencing. Int J Surg 2024; 110:6771-6799. [PMID: 38874485 PMCID: PMC11487005 DOI: 10.1097/js9.0000000000001762] [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: 01/30/2024] [Accepted: 05/24/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Pancreatic cancer, specifically pancreatic ductal adenocarcinoma (PDAC), continues to pose a significant clinical and scientific challenge. The most significant finding of recent years is that PDAC tumours harbour their specific microbiome, which differs amongst tumour entities and is distinct from healthy tissue. This review aims to evaluate and summarise all PDAC studies that have used the next-generation technique, 16S rRNA gene amplicon sequencing within each bodily compartment. As well as establishing a causal relationship between PDAC and the microbiome. MATERIALS AND METHODS This systematic review was carried out according to the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines. A comprehensive search strategy was designed, and 1727 studies were analysed. RESULTS In total, 38 studies were selected for qualitative analysis and summarised significant PDAC bacterial signatures. Despite the growing amount of data provided, we are not able to state a universal 16S rRNA gene microbial signature that can be used for PDAC screening. This is most certainly due to the heterogeneity of the presentation of results, lack of available datasets, and the intrinsic selection bias between studies. CONCLUSION Several key studies have begun to shed light on causality and the influence the microbiome constituents and their produced metabolites could play in tumorigenesis and influencing outcomes. The challenge in this field is to shape the available microbial data into targetable signatures. Making sequenced data readily available is critical, coupled with the coordinated standardisation of data and the need for consensus guidelines in studies investigating the microbiome in PDAC.
Collapse
Affiliation(s)
- Nabeel Merali
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| | - Tarak Chouari
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| | - Casie Sweeney
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
| | - James Halle-Smith
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Maria-Danae Jessel
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| | - Bing Wang
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam
| | - James O’ Brien
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
| | - Satoshi Suyama
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge
| | | | - Keith J. Roberts
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Eirini Velliou
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London (UCL), London
| | - Shivan Sivakumar
- Oncology Department and Institute of Immunology and Immunotherapy, Birmingham Medical School, University of Birmingham
| | - Timothy A. Rockall
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
| | - Ayse Demirkan
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
- Surrey Institute for People-Centred AI, University of Surrey, Guildford, Surrey
| | - Virginia Pedicord
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam
- Fondazione Pisa per la Scienza, San Giuliano, Italy
| | - Nicola E. Annels
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| | - Adam E. Frampton
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| |
Collapse
|
47
|
Mancha S, Horan M, Pasachhe O, Keikhosravi A, Eliceiri KW, Matkowskyj KA, Notbohm J, Skala MC, Campagnola PJ. Multiphoton excited polymerized biomimetic models of collagen fiber morphology to study single cell and collective migration dynamics in pancreatic cancer. Acta Biomater 2024; 187:212-226. [PMID: 39182805 PMCID: PMC11446658 DOI: 10.1016/j.actbio.2024.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/02/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
The respective roles of aligned collagen fiber morphology found in the extracellular matrix (ECM) of pancreatic cancer patients and cellular migration dynamics have been gaining attention because of their connection with increased aggressive phenotypes and poor prognosis. To better understand how collagen fiber morphology influences cell-matrix interactions associated with metastasis, we used Second Harmonic Generation (SHG) images from patient biopsies with Pancreatic ductal adenocarcinoma (PDAC) as models to fabricate collagen scaffolds to investigate processes associated with motility. Using the PDAC BxPC-3 metastatic cell line, we investigated single and collective cell dynamics on scaffolds of varying collagen alignment. Collective or clustered cells grown on the scaffolds with the highest collagen fiber alignment had increased E-cadherin expression and larger focal adhesion sites compared to single cells, consistent with metastatic behavior. Analysis of single cell motility revealed that the dynamics were characterized by random walk on all substrates. However, examining collective motility over different time points showed that the migration was super-diffusive and enhanced on highly aligned fibers, whereas it was hindered and sub-diffusive on un-patterned substrates. This was further supported by the more elongated morphology observed in collectively migrating cells on aligned collagen fibers. Overall, this approach allows the decoupling of single and collective cell behavior as a function of collagen alignment and shows the relative importance of collective cell behavior as well as fiber morphology in PDAC metastasis. We suggest these scaffolds can be used for further investigations of PDAC cell biology. STATEMENT OF SIGNIFICANCE: Pancreatic ductal adenocarcinoma (PDAC) has a high mortality rate, where aligned collagen has been associated with poor prognosis. Biomimetic models representing this architecture are needed to understand complex cellular interactions. The SHG image-based models based on stromal collagen from human biopsies afford the measurements of cell morphology, cadherin and focal adhesion expression as well as detailed motility dynamics. Using a metastatic cell line, we decoupled the roles of single cell and collective cell behavior as well as that arising from aligned collagen. Our data suggests that metastatic characteristics are enhanced by increased collagen alignment and that collective cell behavior is more relevant to metastatic processes. These scaffolds provide new insight in this disease and can be a platform for further experiments such as testing drug efficacy.
Collapse
Affiliation(s)
- Sophie Mancha
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Meghan Horan
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | - Adib Keikhosravi
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kevin W Eliceiri
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI, USA
| | - Kristina A Matkowskyj
- Department of Pathology & Lab Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jacob Notbohm
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Melissa C Skala
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI, USA.
| | - Paul J Campagnola
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
48
|
Liu L, Wang X, Guo D, Ma R, Gong H, Wang C. Hormone replacement therapy and risk of pancreatic cancer in postmenopausal women: Evidence from the US National Inpatient Sample 2008-2018. Heliyon 2024; 10:e37588. [PMID: 39309886 PMCID: PMC11415697 DOI: 10.1016/j.heliyon.2024.e37588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024] Open
Abstract
Background Pancreatic cancer is a serious, usually fatal disease and one of the most aggressive malignancies. Research into whether hormone replacement therapy (HRT) might protect against pancreatic cancer has yielded mixed results. This study aimed to investigate the potential association between HRT and the risk of pancreatic cancer in postmenopausal women. Methods This population-based, retrospective study extracted data from the US National Inpatient Sample (NIS) 2008-2018. Hospitalized females aged ≥55 years were eligible for inclusion. Associations between HRT, other study variables, and pancreatic cancer diagnosis were determined using univariate and multivariable regression analyses. Results After 1:4 matching by age, data of postmenopausal women with (n = 35,309) and without (n = 141,236) HRT were included in the analysis. The mean age was 73.4 years. Multivariable analyses showed that women with HRT had significantly decreased odds of pancreatic cancer (adjusted OR [aOR], 0.69, 95 % CI: 0.53-0.90). Compared to patients without HRT, patients with HRT in the 55-64-year-old group (aOR 0.48, 95 % CI: 0.32-0.74), 65-74-year-old group (aOR 0.49, 95 % CI: 0.34-0.71), non-hypertensive group (aOR 0.55, 95 % CI: 0.38-0.79), and non-hyperlipidemia group (aOR 0.59, 95 % CI: 0.42-0.82) had significantly decreased odds of pancreatic cancer. Conclusions In US postmenopausal women, HRT is associated with a reduced risk of pancreatic cancer, especially those aged 55-74 year. Further study is needed to clarify the mechanisms underlying the associations.
Collapse
Affiliation(s)
- Lei Liu
- Department of Digestive Surgery, Songjiang Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201600, China
| | - Xinyu Wang
- Division of Biology, University of California San Diego, 9500 Gilman Dr. La Jolla, CA, 92093, USA
| | - Dekai Guo
- Department of Digestive Surgery, Songjiang Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201600, China
| | - Ruirui Ma
- Department of Digestive Surgery, Songjiang Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201600, China
| | - Haibing Gong
- Department of Digestive Surgery, Songjiang Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201600, China
| | - Congjun Wang
- Department of Biliary and Pancreatic Surgery, Renji Hospital, Shanghai Jiaotong University, School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| |
Collapse
|
49
|
Lee H, Bae AN, Yang H, Lee JH, Park JH. Modulation of PRC1 Promotes Anticancer Effects in Pancreatic Cancer. Cancers (Basel) 2024; 16:3310. [PMID: 39409930 PMCID: PMC11475828 DOI: 10.3390/cancers16193310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/11/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Pancreatic cancer, while relatively uncommon, is extrapolated to become the second leading cause of cancer-related deaths worldwide. Despite identifying well-known markers like the KRAS gene, the exact regulation of pancreatic cancer progression remains elusive. Methods: Clinical value of PRC1 was analyzed using bioinformatics database. The role of PRC1 was further evaluated through cell-based assays, including viability, wound healing, and sensitivity with the drug. Results: We demonstrate that PRC1 was significantly overexpressed in pancreatic cancer compared to pancreases without cancer, as revealed through human databases and cell lines analysis. Furthermore, high PRC1 expression had a negative correlation with CD4+ T cells, which are crucial for the immune response against cancers. Additionally, PRC1 showed a positive correlation with established pancreatic cancer markers. Silencing PRC1 expression using siRNA significantly inhibited cancer cell proliferation and viability and increased chemotherapeutic drug sensitivity. Conclusions: These findings suggest that targeting PRC1 in pancreatic cancer may enhance immune cell infiltration and inhibit cancer cell proliferation, offering a promising avenue for developing anticancer therapies.
Collapse
Affiliation(s)
| | | | | | | | - Jong Ho Park
- Department of Anatomy, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
50
|
Cho CJ, Nguyen T, Rougeau AK, Huang YZ, To S, Lin X, Gamage ST, Meier JL, Mills JC. Inhibition of Ribosome Biogenesis in vivo Causes p53-Dependent Death and p53-Independent Dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614959. [PMID: 39386693 PMCID: PMC11463434 DOI: 10.1101/2024.09.25.614959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Ribosomes are critical for cell function; their synthesis (known as ribosome biogenesis; "RiBi") is complex and energy-intensive. Surprisingly little is known about RiBi in differentiated cells in vivo in adult tissue. Here, we generated mice with conditional deletion of Nat10 , an essential gene for RiBi and translation, to investigate effects of RiBi blockade in vivo. We focused on RiBi in a long-lived, ribosome-rich cell population, pancreatic acinar cells, during homeostasis and tumorigenesis. We observed a surprising latency of several weeks between Nat10 deletion and onset of structural and functional abnormalities and p53-dependent acinar cell death, which was associated with translocation of ribosomal proteins RPL5 and RPL11 into acinar cell nucleoplasm. Indeed, deletion of Trp53 could rescue acinar cells from apoptotic cell death; however, Nat10 Δ / Δ ; Trp53 Δ / Δ acinar cells remained morphologically and functionally abnormal. Moreover, the deletion of Trp53 did not rescue the lethality of inducible, globally deleted Nat10 in adult mice nor did it rescue embryonic lethality of global Nat10 deletion, emphasizing p53-independent consequences of RiBi inhibition. Deletion of Nat10 in acinar cells blocked Kras -oncogene-driven pancreatic intraepithelial neoplasia and subsequent pancreatic ductal adenocarcinoma, regardless of Trp53 mutation status. Together, our results provide initial insights into how cells respond to defects in RiBi and translation in vivo .
Collapse
|