1
|
Du Nguyen D, Shuklin F, Barulina E, Albitskaya H, Novikov S, Chernov AI, Kim I, Barulin A. Recent advances in dynamic single-molecule analysis platforms for diagnostics: Advantages over bulk assays and miniaturization approaches. Biosens Bioelectron 2025; 278:117361. [PMID: 40117897 DOI: 10.1016/j.bios.2025.117361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/23/2025]
Abstract
Single-molecule science is a unique technique for unraveling molecular biophysical processes. Sensitivity to single molecules provides the capacity for the early diagnosis of low biomarker amounts. Furthermore, the miniaturization of instruments for portable diagnostic tools toward point-of-care testing (POCT) is a crucial development in this field. Herein, we discuss recent developments in single-molecule sensing platforms and their advantages for diagnostics over bulk measurements including molecular size measurements, interaction dynamics, and fast biomarker sensing and sequencing at low concentrations. We highlight the capabilities of dynamic optical and electrical sensing platforms for single-biomolecule and single-vesicle monitoring associated with neurodegenerative disorders, viral diseases, cancers, and more. Current approaches to instrument miniaturization have brought technology closer to portable diagnostics settings via smartphone-based devices, multifunctional portable microscopes, handheld electrical circuit devices, and remote single-molecule assays. Finally, we provide an overview of the clinical applications of single-molecule sensors in POCT assays. Altogether, single-molecule analyses platforms exhibit significant potential for the development of novel portable healthcare devices.
Collapse
Affiliation(s)
- Dang Du Nguyen
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Fedor Shuklin
- Moscow Center for Advanced Studies, Kulakova str. 20, Moscow, 123592, Russia
| | - Elena Barulina
- Moscow Center for Advanced Studies, Kulakova str. 20, Moscow, 123592, Russia; Russian Quantum Center, Moscow, 121205, Russia
| | - Hristina Albitskaya
- Moscow Center for Advanced Studies, Kulakova str. 20, Moscow, 123592, Russia
| | - Sergey Novikov
- Moscow Center for Advanced Studies, Kulakova str. 20, Moscow, 123592, Russia
| | - Alexander I Chernov
- Russian Quantum Center, Moscow, 121205, Russia; Center for Photonics and 2D Materials, Moscow Institute of Physics and Technology, Dolgoprudny, 141700, Russia.
| | - Inki Kim
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Department of MetaBioHealth, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Aleksandr Barulin
- Moscow Center for Advanced Studies, Kulakova str. 20, Moscow, 123592, Russia.
| |
Collapse
|
2
|
Wang L, Jia Y, Wang J, Xia XH, Wang C. Multiplexed Detection of Pancreatic-Specific Nucleic Acids and Protein Biomarkers Using a Logic Nanofluidic Platform. Anal Chem 2025. [PMID: 40387859 DOI: 10.1021/acs.analchem.5c01978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Early detection of pancreatic cancer is vital for patient survival. However, current diagnostic approaches remain constrained by insufficient precision and specificity inherent to single-biomarker detection strategies. Herein, we develop a nanochannel biosensing platform implementing cooperative dual-signal detection of pancreatic-specific biomarkers CA19-9 and miRNA-196a. Using liquid-liquid interface self-assembly, we constructed anodic aluminum oxide (AAO)-Au hybrid nanochannels integrated with a surface-modified double-key DNA nanolock (DDN). The conformational switch of DDN logic gating triggered by miRNA-196a exposes the CA19-9-aptamer, enabling specific target recognition and consequent ion current signal attenuation. Simultaneously, released miRNA-196a is quantified by catalytic hairpin assembly and hybridization chain reaction-mediated cascade amplification. Experiments show that the present DDN-based logic nanofluidic platform could achieve an ultralow detection limit of 0.000027 U·mL-1 for CA19-9 and 4.74 aM for miRNA-196a, which is 2-3 orders of magnitude higher than traditional ELISA/qPCR methods. Finally, clinical sample analysis confirms the high specificity of this platform in distinguishing pancreatic cancer and acute pancreatitis from healthy individuals. This DDN-functionalized nanofluidic biosensor provides valuable insights into designing precision detection platforms for pancreatic cancer, highlighting its significant potential for clinical diagnostics.
Collapse
Affiliation(s)
- Lina Wang
- State Key Laboratory of Microbial Technology, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yixin Jia
- State Key Laboratory of Microbial Technology, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Jin Wang
- State Key Laboratory of Microbial Technology, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Xing-Hua Xia
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Chen Wang
- State Key Laboratory of Microbial Technology, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|
3
|
Zhu Y, Sun R, Fan J, Ma H, Sun B. CSF1-CAR Specifically Targets CSF1R+ Pancreatic Cancer Cells and Tumor-Associated Macrophages. J Immunother 2025:00002371-990000000-00141. [PMID: 40375821 DOI: 10.1097/cji.0000000000000563] [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: 12/10/2024] [Accepted: 04/18/2025] [Indexed: 05/18/2025]
Abstract
SUMMARY A highly suppressive tumor immune microenvironment and nonspecific target endow malignant tumors with CAR-T cells. CSF1R is highly expressed on pancreatic cancer tissues compares with normal tissues in GEPIA database and M2 macrophages mainly contributing to the suppressive tumor microenvironment (TME), suggesting that CSF1R is a suitable antigen. CSF1 is the natural ligand of CSF1R, so we constructed a CSF1-CAR and tested its cytotoxic effect on tumor cells and macrophages in vitro. Our results demonstrated that CSF1-CAR-T cells can lyse tumor cells dependent on CSF1R expression. Meanwhile, CSF1-CAR-T also lyse CSF1R+ M2 macrophages, suggesting that CSF1-CAR-T cells play a role in eliminating tumor cells and remodeling the TME.
Collapse
Affiliation(s)
- Yongjie Zhu
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Cell Engineering and Immunotherapy, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruipu Sun
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Cell Engineering and Immunotherapy, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiawei Fan
- General (specialized) Department, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital& Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Haiyan Ma
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Cell Engineering and Immunotherapy, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi province, P.R. China
| | - Bin Sun
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Cell Engineering and Immunotherapy, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
4
|
Roscigno G, Jacobs S, Toledo B, Borea R, Russo G, Pepe F, Serrano MJ, Calabrò V, Troncone G, Giovannoni R, Giovannetti E, Malapelle U. The potential application of stroma modulation in targeting tumor cells: focus on pancreatic cancer and breast cancer models. Semin Cancer Biol 2025:S1044-579X(25)00060-4. [PMID: 40373890 DOI: 10.1016/j.semcancer.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer development and spreading being considered as "the dark side of the tumor". Within this term tumor cells, immune components, supporting cells, extracellular matrix and a myriad of bioactive molecules that synergistically promote tumor development and therapeutic resistance, are included. Recent findings revealed the profound impacts of TME on cancer development, serving as physical support, critical mediator and biodynamic matrix in cancer evolution, immune modulation, and treatment outcomes. TME targeting strategies built on vasculature, immune checkpoints, and immuno-cell therapies, have paved the way for revolutionary clinical interventions. On this basis, the relevance of pre-clinical and clinical investigations has rapidly become fundamental for implementing novel therapeutical strategies breaking cell-cell and cell -mediators' interactions between TME components and tumor cells. This review summarizes the key players in the breast and pancreatic TME, elucidating the intricate interactions among cancer cells and their essential role for cancer progression and therapeutic resistance. Different tumors such breast and pancreatic cancer have both different and similar stroma features, that might affect therapeutic strategies. Therefore, this review aims to comprehensively evaluate recent findings for refining breast and pancreatic cancer therapies and improve patient prognoses by exploiting the TME's complexity in the next future.
Collapse
Affiliation(s)
- Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy.
| | - Sacha Jacobs
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| | - Belen Toledo
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain.
| | - Roberto Borea
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| | - Gianluca Russo
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Maria Jose Serrano
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy; GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception Group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain.
| | - Viola Calabrò
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Roberto Giovannoni
- Department of Biology, Genetic Unit, University of Pisa, Via Derna 1, 56126 Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy.
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| |
Collapse
|
5
|
Gros B, Alañón Martínez PE, Orti Cuerva M, Aparicio-Serrano A, Gallego Jiménez E, Santos Lucio A, Pleguezuelo Navarro M, Hervás Molina A, Serrano Ruiz FJ. Diagnostic yield of biliary brush cytology via ERCP - A 7-year tertiary center experience. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2025. [PMID: 40353432 DOI: 10.17235/reed.2025.11158/2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
BACKGROUND Biliary brushing cytology during endoscopic retrograde cholangiopancreatography (ERCP) is used to assess the nature of a biliary stricture. Its low sensitivity challenges exclusion of malignancy through this technique. The aim was to evaluate the diagnostic yield of brush cytology in biliary strictures and to identify predictive factors associated with a positive diagnosis of malignancy. METHODS Observational retrospective study in a tertiary center. All adult patients undergoing a biliary brushing during ERPC from 2016 to 2022 were included. Logistic regression analyses were performed to identify predictive factors for positive brush cytology. RESULTS A total of 5309 patients underwent ERCP within the evaluated period. Out of these, biliary brushing was performed in 518 patients including 568 cytology samples, 57.7% (299) were men, median age 74 (64-84) years old. There were 24% (126) benign strictures and 76% (392) malignant of which the most common etiology were pancreatic cancer 42.5% (220/518), followed by cholangiocarcinoma 22.6% (117/518). The sensitivity, specificity, positive predictive value, and negative predictive value were 48%, 98%, 98% and 37%, respectively. Sensitivity was 45% and 52% in pancreatic adenocarcinoma and cholangiocarcinoma, respectively. Older age (OR 1.02, 95% CI: 1.01-1.03, p=0.01) and higher bilirubin (OR 1.05, 95% CI: 1.03-1.08, p<0.001) were independent predictors for brush cytology positivity. There were 9.7% (45/518) post-ERCP complications. CONCLUSIONS Biliary brushing cytology during ERCP is a safe procedure with low sensitivity but high specificity. Older age and higher bilirubin are associated to positive biliary cytology.
Collapse
Affiliation(s)
- Beatriz Gros
- Gastroenterology, Hospital Universitario Reina Sofía, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Pereira-Silva M, Veiga F, Paiva-Santos AC, Concheiro A, Alvarez-Lorenzo C. Biomimetic nanosystems for pancreatic cancer therapy: A review. J Control Release 2025; 383:113824. [PMID: 40348133 DOI: 10.1016/j.jconrel.2025.113824] [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: 12/17/2024] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Pancreatic cancer (PC) is a highly lethal and aggressive malignancy, currently one of the leading causes of cancer-related deaths worldwide, in both women and men. PC is highly resistant to standard chemotherapy (CT) because its immunosuppressive and hypoxic tumor microenvironment and a dense desmoplastic stroma compartment extensively limit drug accessibility and perfusion. Although CT is one of the main therapeutic strategies for PC management contributing to tumor eradication through a cytotoxic effect, CT is associated with a poor pharmacokinetic profile and provokes deleterious systemic toxicity. This low efficacy-poor safety scenario urgently calls for innovative and highly specific therapeutic strategies to counteract this urgent clinical challenge. Nanotechnology-based precision materials for cancer may help improve drug stability and minimize the systemic cytotoxic effects by increasing drug tumor accumulation and also enabling controlled release, but several drawbacks still persist, such as the poor targeting efficiency. In the last few years increased attention has been paid to bioinspired nanosystems that can mimic either partially or totally biological systems, including lipid layers as suitable stealth coatings resembling the composition of cell membranes, lipoprotein- and blood protein-based nanosystems, and cell membrane-derived systems, such as extracellular vesicles, cell membrane nanovesicles and cell membrane-coated nanosystems, which display intrinsic cancer-targeting abilities, enhanced biocompatibility, decreased immunogenicity, and prolonged blood circulation profile. This review covers the recent breakthroughs on advanced biomimetic PC-targeted nanosystems, focusing on their design, properties and applications as innovative, multifunctional and versatile tools paving the way to improved PC diagnosis and treatment.
Collapse
Affiliation(s)
- Miguel Pereira-Silva
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal; Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma, Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma, Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma, Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
7
|
Duhn J, von Fritsch L, Bolm L, Braun R, Honselmann K, Litkevych S, Kist M, Deichmann S, Tol KKV, Franke B, Reinwald F, Sackmann A, Holleczek B, Krauß A, Klinkhammer-Schalke M, Zeissig SR, Keck T, Wellner UF, Abdalla TSA. Perioperative and oncologic outcomes after total pancreatectomy and pancreatoduodenectomy for pancreatic head adenocarcinoma-A propensity score-matched analysis from the German Cancer Registry Group. Surgery 2025; 181:109292. [PMID: 40101369 DOI: 10.1016/j.surg.2025.109292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/08/2025] [Accepted: 02/01/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND To compare perioperative morbidity and mortality in patients receiving pancreatoduodenectomy or total pancreatectomy for pancreatic head adenocarcinoma using German Cancer Registry data. METHODS Anonymized pooled data were retrieved from regional cancer registries participating in the German Cancer Registry Group of the Association of German Tumor Centers. Included were patients diagnosed with pancreatic head adenocarcinoma since 2016, receiving curative intent pancreatoduodenectomy or total pancreatectomy. Patients were propensity-score matched according to age, sex, and histopathology. Primary endpoints were 30- and 90-day postoperative mortality. Secondary endpoints were administration of adjuvant chemotherapy, long-term survival, and patterns of cancer recurrence. The data were analyzed using R. RESULTS In total, 756 patients per treatment group were matched for further analyses. R0-resection rate was comparable between pancreatoduodenectomy and total pancreatectomy (69.6 vs 73.4%, P = .154). The 30-day (9.5 vs 4.8%, P < .001) and 90-day postoperative mortality (18.0 vs 11.0%, P < .001) rates were significantly lower after pancreatoduodenectomy compared with total pancreatectomy. After pancreatoduodenectomy, more patients received adjuvant chemotherapy (43.6 vs 53.3%, P < .001) and time to adjuvant chemotherapy was shorter (60.1 vs 52.7 days, P = .002) compared with total pancreatectomy. Long-term overall survival was worse after total pancreatectomy (P < .001), also in patients receiving adjuvant chemotherapy (P = .019). The sites of recurrence were comparable between both groups (P = .274). CONCLUSION The results of this study show greater perioperative morbidity and mortality after total pancreatectomy compared with pancreatoduodenectomy for pancreatic head malignancy. Also, long-term survival was worse after total pancreatectomy. These results emphasize the role of pancreatoduodenectomy as a standard surgical procedure for pancreatic head adenocarcinoma and suggest that total pancreatectomy should only be performed in selected patients.
Collapse
Affiliation(s)
- Jannis Duhn
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Lennart von Fritsch
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Louisa Bolm
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Rüdiger Braun
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Kim Honselmann
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Stanislav Litkevych
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Markus Kist
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Steffen Deichmann
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Kees Kleihues-van Tol
- Network for Care, Quality and Research in Oncology, German Cancer Registry Group of the Association of German Tumor Centers (ADT), Berlin, Germany
| | - Bianca Franke
- Network for Care, Quality and Research in Oncology, German Cancer Registry Group of the Association of German Tumor Centers (ADT), Berlin, Germany
| | - Fabian Reinwald
- Cancer Registry of Rhineland-Palatinate in the Institute for Digital Health Data, Mainz, Germany
| | - Andrea Sackmann
- Hessian Cancer Registry, Hessian Office for Health and Care, Frankfurt, Germany
| | | | - Anna Krauß
- Cancer Registry Mecklenburg-Western Pomerania, Greifswald, Germany
| | - Monika Klinkhammer-Schalke
- Network for Care, Quality and Research in Oncology, German Cancer Registry Group of the Association of German Tumor Centers (ADT), Berlin, Germany
| | - Sylke R Zeissig
- Network for Care, Quality and Research in Oncology, German Cancer Registry Group of the Association of German Tumor Centers (ADT), Berlin, Germany; Institute of Clinical Epidemiology and Biometry (ICE-B), University of Würzburg, Würzburg, Germany
| | - Tobias Keck
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.
| | - Ulrich F Wellner
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Thaer S A Abdalla
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| |
Collapse
|
8
|
Hu C, Liu H, Zhang Z, Li L, Mao GJ, Cheng W, Zhou L. A Self-Calibrating Fluorescent-Photoacoustic Integrated Probe Enables Fast Visualizing Pancreatic Cancer and Imaging-Guided Tumor Surgery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408527. [PMID: 39593243 DOI: 10.1002/smll.202408527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/03/2024] [Indexed: 11/28/2024]
Abstract
Pancreatic cancer is known for its high invasiveness and metastasis, making rapid visualization and precise treatment critical for improving patient outcomes. Current diagnostic tools lack abilities to provide rapid and accurate tumor localization, particularly for real-time intraoperative guidance. To address this gap, the study has developed a novel Förster Resonance Energy Transfer (FRET)-mediated dual-ratiometric near-infrared fluorescence (NIRF)/photoacoustic (PA) bimodal probe, SiRho-SHD-NTR, specifically designed for the fast and accurate navigation of pancreatic tumor resection. The probe, due to its excellent binding affinity with nitroreductase (NTR), can rapidly reach response saturation. Cellular experiments demonstrate that the probe rapidly and efficiently penetrates cancer cells, enhancing the effectiveness of PA imaging for preliminary diagnosis and tumor localization, while also enabling the rapid visualization of pancreatic tumors through NIRF imaging. Leveraging the rapid response characteristics of the probe to NTR, the study achieves precise tumor imaging in orthotopic pancreatic cancer mice by spraying the probe, within ≈5 min. More importantly, the probe even allows for the fast visualization of metastatic tumors and fluorescence-guided surgical resection. It is believed that SiRho-SHD-NTR will offer a promising method in the rapid visualization of pancreatic cancer and provide a powerful tool for imaging-guided tumor surgery, targeting both primary and metastatic tumors.
Collapse
Affiliation(s)
- Cong Hu
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, P. R. China
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, P. R. China
| | - Hongwen Liu
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, P. R. China
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, P. R. China
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, P. R. China
| | - Zhipengjun Zhang
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, P. R. China
| | - Lingyun Li
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, P. R. China
| | - Guo-Jiang Mao
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, 453007, P. R. China
| | - Wei Cheng
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, P. R. China
| | - Lei Zhou
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, P. R. China
| |
Collapse
|
9
|
Ramai D, Collins B, Ofosu A, Mohan BP, Jagannath S, Tabibian JH, Girotra M, Barakat MT. Deep Learning Methods in the Imaging of Hepatic and Pancreaticobiliary Diseases. J Clin Gastroenterol 2025; 59:405-411. [PMID: 40193287 DOI: 10.1097/mcg.0000000000002125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Reports indicate a growing role for artificial intelligence (AI) in the evaluation of pancreaticobiliary and hepatic conditions. A key focus is differentiating between benign and malignant lesions, which is crucial for treatment decisions. AI improves diagnostic accuracy through high sensitivity and specificity, while CNN algorithms enhance image analysis and reduce variability. These advancements aim to match the accuracy of pathologists in cancer detection. In addition, AI aids in identifying diagnostic markers, as early detection is essential. This article reviews the applications of machine learning and deep learning in the diagnosis of hepatic and pancreaticobiliary diseases. Although the use of AI in these specialized areas of gastroenterology is primarily confined to experimental trials, current models demonstrate significant potential for enhancing the detection, evaluation, and treatment planning of hepatic and pancreaticobiliary conditions.
Collapse
Affiliation(s)
- Daryl Ramai
- Division of Gastroenterology and Hepatology, University of Utah, Salt Lake City, UT
| | - Brendan Collins
- Division of Digestive Diseases, University of Cincinnati, Cincinnati, OH
| | - Andrew Ofosu
- Division of Digestive Diseases, University of Cincinnati, Cincinnati, OH
| | - Babu P Mohan
- Division of Gastroenterology and Hepatology, University of Utah, Salt Lake City, UT
| | - Soumya Jagannath
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - James H Tabibian
- Division of Gastroenterology, Olive View-UCLA Medical Center, Sylmar
- David Geffen School of Medicine at UCLA, Los Angeles
| | - Mohit Girotra
- Digestive Health Institute, Swedish Medical Center, Seattle, WA
| | | |
Collapse
|
10
|
Azeez A, Noel C. Global status of research on quality of life in pancreatic cancer patients: A bibliometric and network analysis from 2005-2024. Clin Res Hepatol Gastroenterol 2025; 49:102595. [PMID: 40210107 DOI: 10.1016/j.clinre.2025.102595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Pancreatic cancer (PC) is a major global health challenge, with rising incidence and mortality rates, particularly in high-socio-demographic index regions. Given its high mortality and significant morbidity, research on patient quality of life (QoL) has gained momentum, addressing symptom burdens, psychological distress, and treatment-related outcomes. Bibliometric analysis provides a valuable approach to mapping research trends, identifying key contributors, and forecasting future directions. OBJECTIVE This study aimed to map global research on QoL in pancreatic cancer patients, highlighting key findings, challenges, and future directions through bibliometric analysis over the past two decades. METHODS Data for this study were collected from the Web of Science Core Collection (WoSCC) database, using specific search strategies to retrieve relevant documents on the quality of life in pancreatic cancer patients. The data were analysed using the Bibliometrix R package to create knowledge maps and visualize research trends, collaborations, and emerging hotspots in the field. RESULTS A total of 819 articles on pancreatic cancer and quality of life were identified, with an average citation count of 47.13 per article, highlighting moderate academic impact. The research revealed a growing trend in collaborative efforts, with an average of 9.42 co-authors per article and 16 % international collaborations. The United States emerged as the leading contributor, with 203 publications and the highest citation count, followed by France and the United Kingdom. CONCLUSION This bibliometric analysis highlights the growing volume of pancreatic cancer and quality of life research, with a steady annual growth rate of 6.9 % and increasing collaboration, especially from the United States. However, despite the rising number of publications, a decline in citation impact for recent studies suggests a need for continued innovation in therapeutic strategies to improve clinical outcomes.
Collapse
Affiliation(s)
- Adeboye Azeez
- Gastrointestinal Research Unit, Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa.
| | - Colin Noel
- Gastrointestinal Research Unit, Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| |
Collapse
|
11
|
Arseneau RJ, Kempster E, Bekkers C, Samson T, Gala-Lopez BL, Ramjeesingh R, Boudreau JE, Arnason T. Claudin 18 (43-14A clone) expression in pancreatic ductal adenocarcinoma: Assessment of a potential clinical biomarker for zolbetuximab therapy. Transl Oncol 2025; 55:102362. [PMID: 40117781 PMCID: PMC11979426 DOI: 10.1016/j.tranon.2025.102362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/08/2025] [Accepted: 03/16/2025] [Indexed: 03/23/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains highly lethal, with a five-year survival rate below 15 %. Claudin 18.2 (CLDN18.2) has emerged as a novel potential therapeutic target in PDAC. Zolbetuximab, a monoclonal antibody targeting CLDN18.2, has demonstrated therapeutic benefit in gastric cancers and is now in phase 2 clinical trials for PDAC. Trial eligibility for zolbetuximab requires tumor membranous immunohistochemical staining with the pan-claudin 18 companion diagnostic antibody clone 43-14A. However, the expression of CLDN18 detected using this clone has only been evaluated in 62 patients from a single retrospective study. Herein, we report immunohistochemical staining using 43-14A on surgically resected PDAC samples (n = 120). Samples were stained following the protocol used in clinical trials, using the 43-14A VENTANA antibody in a prediluted kit, and according to the manufacturer's recommended protocol. Positive cases were defined as ≥ 75 % of tumor cells exhibiting membranous staining with an intensity of ≥ 2+. Out of 120 PDAC cases, 39 (32.5 %) stained positive for CLDN18 with 43-14A. A significant association was observed between lower tumor grade and higher 43-14A staining (p < 0.05). CLDN18-positive cases demonstrated significantly improved survival at the cohort's median overall survival (23 months, p < 0.05), suggesting that claudin expression could serve as a both a diagnostic and prognostic marker. Our findings indicate that 32.5 % of PDAC tumors in this cohort are positive for CLDN18, suggesting that a significant proportion of patients with PDAC could benefit from zolbetuximab and other CLDN18.2 targeted immunotherapies if pancreatic cancer therapeutic trials prove successful.
Collapse
Affiliation(s)
- Riley J Arseneau
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | | | | | | | - Boris L Gala-Lopez
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada; Nova Scotia Health, Halifax, NS, Canada; Department of Microbiology & Immunology, Dalhousie University, NS, Canada
| | - Ravi Ramjeesingh
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada; Nova Scotia Health, Halifax, NS, Canada
| | - Jeanette E Boudreau
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada; Nova Scotia Health, Halifax, NS, Canada; Department of Microbiology & Immunology, Dalhousie University, NS, Canada
| | - Thomas Arnason
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Nova Scotia Health, Halifax, NS, Canada.
| |
Collapse
|
12
|
He W, Cui J, Wang XY, Siu RHP, Tanner JA. Early-Stage Pancreatic Cancer Diagnosis: Serum Biomarkers and the Potential for Aptamer-Based Biosensors. Molecules 2025; 30:2012. [PMID: 40363817 PMCID: PMC12073606 DOI: 10.3390/molecules30092012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Pancreatic cancer has a high mortality rate, and both the incidence and mortality are continuing to increase in many countries globally. The poor prognosis of pancreatic cancer is in part due to the challenges in early diagnosis. Improving early-stage pancreatic cancer diagnosis would improve survival outcomes. Aptamer-based biosensors provide an alternative technological approach for the analysis of serum biomarkers with several potential advantages. This review summarizes the major pancreatic cancer serum biomarkers, as well as discusses recent progress in biomarker exploration and aptasensor development. Here, we review both established and novel serum biomarkers identified recently, emphasizing their potential for early-stage pancreatic cancer diagnosis. We also propose strategies for further expanding multiplex biomarker panels beyond the established CA19-9 biomarker to enhance diagnostic performance. We discuss technological advancements in aptamer-based sensors for pancreatic cancer-related biomarkers over the last decade. Optical and electrochemical sensors are highlighted as two primary modalities in aptasensor design, each offering unique advantages. Finally, we propose steps towards clinical application using aptamer-based sensors with multiplexed biomarker detection for improved pancreatic cancer diagnostics.
Collapse
Affiliation(s)
- Weisi He
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (W.H.); (J.C.); (X.-Y.W.); (R.H.P.S.)
| | - Jingyu Cui
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (W.H.); (J.C.); (X.-Y.W.); (R.H.P.S.)
| | - Xue-Yan Wang
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (W.H.); (J.C.); (X.-Y.W.); (R.H.P.S.)
| | - Ryan H. P. Siu
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (W.H.); (J.C.); (X.-Y.W.); (R.H.P.S.)
| | - Julian A. Tanner
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (W.H.); (J.C.); (X.-Y.W.); (R.H.P.S.)
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Hong Kong SAR, China
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen 518057, China
| |
Collapse
|
13
|
Zhang R, Danshiitsoodol N, Noda M, Yonezawa S, Kanno K, Sugiyama M. Stevia Leaf Extract Fermented with Plant-Derived Lactobacillus plantarum SN13T Displays Anticancer Activity to Pancreatic Cancer PANC-1 Cell Line. Int J Mol Sci 2025; 26:4186. [PMID: 40362423 PMCID: PMC12071683 DOI: 10.3390/ijms26094186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Pancreatic cancer is a highly malignant tumor that remains a significant global health burden. In this study, we demonstrated the anticancer potential of stevia leaf extract fermented with plant-derived Lactobacillus (L.) plantarum SN13T strain. Evaluation of antioxidant capacity (including DPPH and ABTS radical scavenging activities and H2O2-induced oxidative damage repair in HEK-293 cells), as well as cytotoxicity against pancreatic cancer cells (PANC-1) and non-cancerous human embryonic kidney (HEK-293), revealed that the fermented extract exhibited significantly enhanced antioxidant activity and cytotoxicity against PANC-1 cells while showing minimal toxicity to HEK-293 cells compared to the unfermented extract. Further, validation through clonogenic, migration, and wound-healing assays demonstrated that the fermented extract effectively inhibited the proliferation and migration of PANC-1 cells. The active compound in the fermented extract has been identified as chlorogenic acid methyl ester (CAME), with a concentration of 374.4 μg/mL. Flow cytometry analysis indicated that CAME significantly arrested PANC-1 cells in the G0/G1 phase and induced apoptosis. Furthermore, CAME upregulated the expression of pro-apoptotic genes Bax, Bad, Caspase-3/9, Cytochrome c, and E-cadherin, while downregulating the anti-apoptotic gene Bcl-2. These findings suggest that CAME exerts potent cytotoxic effects on PANC-1 cells by inhibiting cell proliferation and migration, arresting the cell cycle, and regulating apoptosis-related gene expression. In conclusion, stevia leaf extract fermented with L. plantarum SN13T, which contains CAME, may serve as a promising candidate for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Rentao Zhang
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (R.Z.); (N.D.); (M.N.)
| | - Narandalai Danshiitsoodol
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (R.Z.); (N.D.); (M.N.)
| | - Masafumi Noda
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (R.Z.); (N.D.); (M.N.)
| | - Sayaka Yonezawa
- Department of General Internal Medicine, Hiroshima University Hospital, Hiroshima University, Hiroshima 734-8551, Japan; (S.Y.); (K.K.)
| | - Keishi Kanno
- Department of General Internal Medicine, Hiroshima University Hospital, Hiroshima University, Hiroshima 734-8551, Japan; (S.Y.); (K.K.)
| | - Masanori Sugiyama
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (R.Z.); (N.D.); (M.N.)
| |
Collapse
|
14
|
Fan H, Zhao H, Gao L, Dong Y, Zhang P, Yu P, Ji Y, Chen ZS, Liang X, Chen Y. CCN1 Enhances Tumor Immunosuppression through Collagen-Mediated Chemokine Secretion in Pancreatic Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500589. [PMID: 40287974 DOI: 10.1002/advs.202500589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/03/2025] [Indexed: 04/29/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense, immunosuppressive tumor microenvironment (TME) that limits therapeutic efficacy. This study investigates the role of cellular communication network factor 1 (CCN1, also known as Cyr61), an extracellular matrix-associated protein, in modulating the TME of PDAC. It is demonstrated that Ccn1 promotes PDAC progression by upregulating collagen and chemokine expression, thereby facilitating immune cell exclusion and enhancing tumor growth. Using a Ccn1-deficient PDAC model, decreased collagen and chemokine levels are observed, resulting in increased infiltration of cytotoxic immune cells and reduced myeloid-derived suppressor cells (MDSCs). Furthermore, Ccn1-deficient tumors exhibit heightened sensitivity to gemcitabine and show enhanced responsiveness to anti-programmed cell death 1 (anti-PD1) therapy. Mechanistically, Ccn1 regulates chemokine production through collagen expression, with chemokine levels remaining suppressed even upon interferon-gamma treatment in collagen-deficient cells. These findings highlight Ccn1 as a potential therapeutic target that reprograms the TME to enhance the efficacy of both chemotherapy and immunotherapy in PDAC, providing a novel approach for overcoming immune resistance in PDAC.
Collapse
Affiliation(s)
- Hongjie Fan
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Huzi Zhao
- Department of Pathology, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, China
| | - Lili Gao
- Department of Pathology, Xinhua Hospital Affiliated to Medicine School of Shanghai Jiaotong University, Shanghai, 200082, China
| | - Yucheng Dong
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100006, China
| | - Pei Zhang
- Department of Mathematics, University of Maryland, College Park, Maryland, MD 20742, USA
| | - Pengfei Yu
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Yunfei Ji
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Xinmiao Liang
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Yang Chen
- State Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| |
Collapse
|
15
|
Zhao B, Fang R, Schürmann H, Hemmer EJ, Mayer GL, Trajkovic-Arsic M, Althoff K, Yang J, Godfrey L, Liffers ST, Savvatakis K, Dorsch M, Grüner BM, Hahn S, Remke M, Lueong SS, Siveke JT. PLK1 blockade enhances the anti-tumor effect of MAPK inhibition in pancreatic ductal adenocarcinoma. Cell Rep 2025; 44:115541. [PMID: 40188436 DOI: 10.1016/j.celrep.2025.115541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/04/2025] [Accepted: 03/19/2025] [Indexed: 04/08/2025] Open
Abstract
Despite constitutive Ras/Raf/MAPK pathway activation in most pancreatic ductal adenocarcinomas (PDACs), treatment approaches targeting this pathway have primarily been unsuccessful. We conduct a drug library screen on an MEK inhibitor (MEKi)-resistant PDAC model and perform complementary pathway analysis to identify cellular resistance phenotypes. We use syngeneic models to investigate the molecular determinants of identified drug synergism. Our study reveals an enrichment for the hallmarks of G2/M checkpoints in MEKi-resistant phenotypes from all investigated models. We find overexpression of Polo-like kinase 1 (PLK1) and other G2/M checkpoint-related proteins in MEKi-resistant cells. We identify synergistic activity between MEK and PLK1 inhibition both in vitro and in vivo and mechanistically show that dual inhibition of the PLK1 and MEK pathways activates the JNK/c-JUN pathway. This causes the accumulation of DNA damage, ultimately leading to apoptotic cell death. Dual PLK1/MEK inhibition emerges as a promising targeted approach in PDAC.
Collapse
Affiliation(s)
- Ben Zhao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Rui Fang
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Hendrik Schürmann
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany; Department of Medical Oncology, West German Cancer Center, University Hospital Essen, 45147 Essen, Germany
| | - Erik Jan Hemmer
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Gina Lauren Mayer
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Marija Trajkovic-Arsic
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Kristina Althoff
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Jiajin Yang
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Laura Godfrey
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Sven T Liffers
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Konstantinos Savvatakis
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany
| | - Madeleine Dorsch
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany; Department of Medical Oncology, West German Cancer Center, University Hospital Essen, 45147 Essen, Germany
| | - Barbara M Grüner
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany; Department of Medical Oncology, West German Cancer Center, University Hospital Essen, 45147 Essen, Germany
| | - Stephan Hahn
- Department of Molecular GI Oncology, Faculty of Medicine, Ruhr University Bochum, 44780 Bochum, Germany; Department of Internal Medicine, Ruhr University Bochum, Knappschaftskrankenhaus, 44780 Bochum, Germany
| | - Marc Remke
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, 40225 Düsseldorf, Germany; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada; Department of Pediatric Neuro-Oncogenomics, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 40225 Düsseldorf, Germany
| | - Smiths S Lueong
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany.
| | - Jens T Siveke
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147 Essen, Germany.
| |
Collapse
|
16
|
Zhou Y, Wu Z, Zeng L, Chen R. Combining genetic and non-genetic factors to predict the risk of pancreatic cancer in patients with new-onset diabetes mellitus. BMC Med 2025; 23:224. [PMID: 40234846 PMCID: PMC12001390 DOI: 10.1186/s12916-025-04048-4] [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: 04/15/2024] [Accepted: 04/02/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Recent research suggests that new-onset diabetes mellitus (NODM) often results from pancreatic cancer (PC) rather than causing it. Determining if NODM is type 2 diabetes mellitus (T2DM) or PC-related NODM (NODM-PC) aids in the early diagnosis of PC. We developed a NODM-PC risk prediction model to stratify PC risk in patients with NODM. METHODS This study utilized data from the UK Biobank, including 238 NODM-PC cases and 14,825 cancer-free T2DM controls. Polygenic risk scores (PRSs) for PC and T2DM were constructed using previously reported single nucleotide polymorphisms (SNPs) separately, while the NODM-PC PRS was developed by combining SNPs from both. Non-genetic factors were selected as candidate predictors based on prior NODM-PC prediction models. We developed three Cox models to estimate the risk of PC diagnosis within 3 years in the NODM population and evaluated them by internal-external cross-validation. RESULTS Elevated NODM-PC PRS and PC PRS scores positively correlated with NODM-PC risk, while T2DM PRS showed an inverse correlation. The NODM-PC PRS achieved the highest AUC at 0.719. Three Cox models were developed: Model 1 included age at T2DM diagnosis, smoking status, HbA1c, PC PRS, and T2DM PRS; Model 2 replaced PC and T2DM PRS with NODM-PC PRS; Model 3 included only non-genetic factors. Model 2 had the highest discrimination (Harrell's C-index 0.823 (95% CI: 0.806-0.840)), demonstrated the best clinical utility with good calibration, and showed significant classification improvement (continuous net reclassification index: 26.89% and 31.93% for cases, 28.51% and 30.90% for controls, compared to Models 1 and 3). The positive predictive value for the top 1% predicted risk in Model 2 was 13.25%. CONCLUSIONS This NODM-PC PRS enhances NODM-PC risk prediction, efficiently identifies individuals at high risk for PC screening, and improves PC screening efficiency at the population level among NODM individuals.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhuo Wu
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Liangtang Zeng
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China.
| | - Rufu Chen
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China.
| |
Collapse
|
17
|
Byeon S, Abbott B, Roach P, Bailey DL, Chou A, Maloney S, Gill AJ, Samra J, Mittal A, Sahni S. Total lesion glycolysis is a promising predictor of chemo-response in pancreatic cancer patients treated with neoadjuvant chemotherapy prior to surgery. Eur J Clin Invest 2025:e70046. [PMID: 40197984 DOI: 10.1111/eci.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/29/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND There has been increased use of neoadjuvant chemotherapy (NAC) in resectable pancreatic ductal adenocarcinoma (PDAC) patients. [18F]fluoro-2-deoxyglucose positron emission tomography/computed tomography (FDG-PET/CT) scan is being frequently used to determine treatment response in PDAC patients undergoing NAC. Maximum standardized uptake value (SUVmax) is conventionally used as an FDG-PET/CT parameter, but there are emerging parameters, such as total lesion glycolysis (TLG), which take into account mean standardized uptake (SUVmean) and metabolic tumour volume (MTV). This study compared the ability of emerging FDG-PET/CT parameters (i.e. SUVmean, MTV and TLG) to predict chemo-response compared to SUVmax. METHODS In this single centre, retrospective study, NAC-treated PDAC patients (n = 74) for whom both pre- and post-NAC FDG-PET/CT scans were available were recruited. All scans were imported to a single analysis platform and reanalysed. Chemo-response was determined by the assessment of percentage viable tumour cells in the tumour bed. Statistical analysis was performed on the data. RESULTS A significant correlation was observed between post-treatment FDG-PET/CT scan parameters and viable cancer cells in the tumour bed, with TLG showing a higher degree of correlation (r = .3131) compared to all other parameters (r = .2722-.3008). The percentage decrease in the TLG (post-NAC scan vs. pre-NAC scan) demonstrated the highest degree of correlation with viable cancer cells in the tumour bed (r = -.3444) and had a statistically significant (p = .0157) effect between NAC responders (Median = 80.57) and non-responders (Median = 65.16). The difference between TLG (post-NAC scan vs. pre-NAC scan) was shown to be an independent prognostic indicator for overall survival (hazard ratio = .5033, p = .0361). CONCLUSION TLG was shown to be a superior predictor of chemo-response and patient prognosis compared to all other FDG-PET/CT parameters in PDAC patients treated with NAC.
Collapse
Affiliation(s)
- Sooin Byeon
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards, New South Wales, Australia
- Kolling Institute of Medical Research, University of Sydney, St Leonards, New South Wales, Australia
| | - Bridget Abbott
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Paul Roach
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Dale L Bailey
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards, New South Wales, Australia
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Angela Chou
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards, New South Wales, Australia
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Sarah Maloney
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards, New South Wales, Australia
- Kolling Institute of Medical Research, University of Sydney, St Leonards, New South Wales, Australia
| | - Anthony J Gill
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards, New South Wales, Australia
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Jaswinder Samra
- Australian Pancreatic Centre, St Leonards, Sydney, New South Wales, Australia
- Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, St Leonards, New South Wales, Australia
| | - Anubhav Mittal
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards, New South Wales, Australia
- Australian Pancreatic Centre, St Leonards, Sydney, New South Wales, Australia
- Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, St Leonards, New South Wales, Australia
- The University of Notre Dame Australia, Sydney, New South Wales, Australia
| | - Sumit Sahni
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards, New South Wales, Australia
- Kolling Institute of Medical Research, University of Sydney, St Leonards, New South Wales, Australia
| |
Collapse
|
18
|
Liaki V, Rosas-Perez B, Guerra C. Unlocking the Genetic Secrets of Pancreatic Cancer: KRAS Allelic Imbalances in Tumor Evolution. Cancers (Basel) 2025; 17:1226. [PMID: 40227826 PMCID: PMC11987834 DOI: 10.3390/cancers17071226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/13/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) belongs to the types of cancer with the highest lethality. It is also remarkably chemoresistant to the few available cytotoxic therapeutic options. PDAC is characterized by limited mutational heterogeneity of the known driver genes, KRAS, CDKN2A, TP53, and SMAD4, observed in both early-stage and advanced tumors. In this review, we summarize the two proposed models of genetic evolution of pancreatic cancer. The gradual or stepwise accumulated mutations model has been widely studied. On the contrary, less evidence exists on the more recent simultaneous model, according to which rapid tumor evolution is driven by the concurrent accumulation of genetic alterations. In both models, oncogenic KRAS mutations are the main initiating event. Here, we analyze the emerging topic of KRAS allelic imbalances and how it arises during tumor evolution, as it is often detected in advanced and metastatic PDAC. We also summarize recent evidence on how it affects tumor biology, metastasis, and response to therapy. To this extent, we highlight the necessity to include studies of KRAS allelic frequencies in the design of future therapeutic strategies against pancreatic cancer.
Collapse
Affiliation(s)
- Vasiliki Liaki
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
| | - Blanca Rosas-Perez
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
| | - Carmen Guerra
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
19
|
Ren J, Su T, Ding J, Chen F, Mo J, Li J, Wang Z, Han L, Wu Z, Wu S. Chlorophyllin exerts synergistic anti-tumor effect with gemcitabine in pancreatic cancer by inducing cuproptosis. Mol Med 2025; 31:126. [PMID: 40186145 PMCID: PMC11969790 DOI: 10.1186/s10020-025-01180-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
Pancreatic cancer (PC) has high lethality due to multiple reasons, and its limited response to conventional chemotherapy like gemcitabine (GEM) is a non-negligible one. Therefore, our study introduces Chlorophyllin (CHL) as an effective therapeutic candidate to enhance the therapeutic efficacy of GEM. Our results demonstrate that the combination of CHL and GEM exhibits a significant synergistic anti-tumor effect by targeting multiple oncogenic processes in PC, including inhibiting cell proliferation, invasion, and migration, as well as inducing cell apoptosis. Further investigations of mechanism have revealed that CHL induces cuproptosis in PC cells through a multifaceted process, involving depleting cellular intracellular glutathione (GSH), increasing reactive oxygen species (ROS) levels, and subsequently upregulating the HSP70 protein in response to heightened oxidative stress. Additionally, CHL releases free Cu2+, binds to the Ferredoxin 1 (FDX1) protein, and ultimately leads to the oligomerization of Dihydrolipoamide S-Acetyltransferase (DLAT) proteins to amplify the copper toxicity within PC cells. Moreover, in vivo experiments have demonstrated that the combination of CHL and GEM effectively inhibits the growth of subcutaneously transplanted tumors while maintaining a favorable biosafety profile. In conclusion, our study identifies CHL as a potent enhancer of GEM's anti-tumor effects in PC through the induction of cuproptosis, thus providing a novel therapeutic avenue for patients with PC.
Collapse
Affiliation(s)
- Jiaqiang Ren
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Tong Su
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Jiachun Ding
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Fan Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jiantao Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jie Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Liang Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Shuai Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
20
|
McMorrow R, de Bruijn HS, Farina S, van Ardenne RJ, Que I, Mastroberardino PG, Robinson DJ, Mezzanotte L, Löwik CW. Combination of Bremachlorin PDT and Immune Checkpoint Inhibitor Anti-PD-1 Shows Response in Murine Immunological T-cell-High and T-cell-Low PDAC Models. Mol Cancer Ther 2025; 24:605-617. [PMID: 39704624 PMCID: PMC11962392 DOI: 10.1158/1535-7163.mct-23-0733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 04/12/2024] [Accepted: 12/17/2024] [Indexed: 12/21/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most challenging types of cancer with little or no response to immune checkpoint inhibitors (ICI). Photodynamic therapy (PDT) has been shown to ablate tumors and induce an immune response. In our study, we investigated the effect of PDT using the photosensitizer Bremachlorin, in its ability to reduce tumor burden and immunologically sensitize T-cell-high and T-cell-low murine PDAC tumors to the ICIs that blocks PD-1 immune checkpoint. In addition, we monitored the effect on survival and investigated if there was a response in PDT-treated and non-PDT-treated distant tumors. Our results showed that Bremachlorin PDT induces direct tumor killing that increased survival in both "hot" T-cell-high and "cold" T-cell-low PDAC tumors and that it can make T-cell-high tumors more sensitive to ICIs blocking PD-1. We found that T-cell-high tumor-bearing mice had an overall greater response to therapy than did T-cell-low tumor-bearing mice. One mouse with T-cell-high tumors exhibited complete tumor regression in both the treated and nontreated distant tumor 90 days after treatment. These results indicate that combining ICIs with Bremachlorin PDT could be a promising therapeutic intervention for enhancing PDAC's response to therapy.
Collapse
Affiliation(s)
- Roisin McMorrow
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Henriette S. de Bruijn
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Stefania Farina
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Ruben J.L. van Ardenne
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Ivo Que
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Pier G. Mastroberardino
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
- IFOM-The FIRC Institute of Molecular Oncology, Milan, Italy
- Department of Life, Health, and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Dominic J. Robinson
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Laura Mezzanotte
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
- Department of Molecular Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Clemens W.G.M. Löwik
- Department of Radiology and Nuclear Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| |
Collapse
|
21
|
Li M, Li T, Liu Y, Han D, Wu S, Gong J. Dual Cascade-Responsive Multifunctional Nanoparticles to Overcome Bacterium-Induced Drug Inactivation and Enhanced Photodynamic and Chemo-Immunotherapy of Pancreatic Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412707. [PMID: 40095308 DOI: 10.1002/smll.202412707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/16/2025] [Indexed: 03/19/2025]
Abstract
The harsh biological barriers and bacteria within tumor microenvironment not only hinder drug penetration and induce drug inactivation, but also inhibit antitumor immune responses. Here a tumor microenvironment dual cascade-responsive multifunctional nanoparticle, Gem/Emo@NP@GHA is reported, which is engineered from a hyaluronidase (HAase)-responsive guanidine group functionalized hyaluronic acid (GHA) shell and a glutathione (GSH)-responsive biopolymer core (Gem/Emo@NP), that encapsulates anticancer drug gemcitabine (Gem) and two-photon-excited photosensitizer emodin (Emo). The constructed Gem/Emo@NP@GHA can specifically target the tumor and subsequently be degraded by HAase-abundant in the extracellular matrix. Thus, the resulting Gem/Emo@NP achieved size reduction and charge reversal, strengthening deep tumor penetration. Upon internalization, the positively charged Gem/Emo@NP effectively kills intratumor bacteria by inducing membrane depolarization. Furthermore, the high levels of GSH within tumor cells disrupt the disulfide bonds of Gem/Emo@NP, triggering drug release. Thereby, the undecomposed Gem successfully induces tumor cell apoptosis and necrosis. Under laser irradiation, photosensitizer Emo generates high singlet oxygen (1O2), further eliminating tumors and intracellular bacteria. More importantly, Gem/Emo@NP@GHA can activate T cell-mediated immune response, further enhancing antitumor activity. These findings provide a promising approach to treating bacterially infected tumors through the synergistic application of chem-immunotherapy and two-photon-excited photodynamic therapy.
Collapse
Affiliation(s)
- Maolin Li
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin, 300072, P. R. China
| | - Tong Li
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin, 300072, P. R. China
| | - Yin Liu
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, P. R. China
| | - Dandan Han
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin, 300072, P. R. China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, 300192, P. R. China
| | - Songgu Wu
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin, 300072, P. R. China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, 300192, P. R. China
| | - Junbo Gong
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin, 300072, P. R. China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, 300192, P. R. China
| |
Collapse
|
22
|
Liu Y, Li C, Cui X, Li M, Liu S, Wang Z. Potentially diagnostic and prognostic roles of piRNAs/PIWIs in pancreatic cancer: A review. Biochim Biophys Acta Rev Cancer 2025; 1880:189286. [PMID: 39952623 DOI: 10.1016/j.bbcan.2025.189286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with limited early diagnostic methods and therapeutic options, contributing to its poor prognosis. Recent advances in high-throughput sequencing have highlighted the critical roles of noncoding RNAs (ncRNAs), particularly PIWI-interacting RNAs (piRNAs), in cancer biology. In this review, we systematically summarize the emerging roles of piRNAs and their associated PIWI proteins in PDAC pathogenesis, progression, and prognosis. We provide a comprehensive analysis of the molecular mechanisms by which piRNAs/PIWIs regulate gene expression and cellular signaling pathways in PDAC. Furthermore, we discuss their potential as novel biomarkers for early diagnosis and therapeutic targets. Importantly, this review identifies key piRNAs/PIWIs involved in PDAC and proposes innovative strategies for improving diagnosis and treatment outcomes. Our work not only consolidates current knowledge but also offers new perspectives for future research and clinical applications in PDAC management.
Collapse
Affiliation(s)
- Yukun Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changlei Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotong Cui
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Miaomiao Li
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China
| | - Shiguo Liu
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China.
| | - Zusen Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
23
|
Wu Y, Qiao Y, Yang C, Chen Y, Shen X, Deng C, Yao Q, Sun N. Accelerated Exosomal Metabolic Profiling Enabled by Robust On-Target Array Sintering with Metal-Organic Frameworks. SMALL METHODS 2025; 9:e2401238. [PMID: 39263996 DOI: 10.1002/smtd.202401238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/06/2024] [Indexed: 09/13/2024]
Abstract
Pancreatic cancer is highly lethal, and survival chances improve only with early detection at a precancerous stage. However, there remains a significant gap in developing tools for large-scale, rapid screening. To this end, a high-throughput On-Target Array Extraction Platform (OTAEP) by direct sintering of a series of metal-organic frameworks (MOFs) for dual in situ extraction, encompassing both exosomes and their metabolic profiles, is developed. Based on the principle of geometry-dependent photothermal conversion efficiency and standard testing, the appropriate MOF functional unit is identified. This unit enables exosome enrichment within 10 min and metabolic fingerprint extraction in under 1 s of laser irradiation, with over five reuse. To further accelerate and enhance the quality of metabolic profile analysis, the application of Surrogate Variable Analysis to eliminate hidden confounding factors within the profiles is proposed, and five biomarkers demonstrated by MS/MS experiments are identified. These biomarkers enable early diagnosis, risk stratification, and staging of pancreatic cancer simultaneously, with sensitivity of 94.1%, specificity of 98.8%, and precision of 94.9%. This work represents a breakthrough for overcoming throughput challenges in large-scale testing and for addressing confounding factors in big data analysis.
Collapse
Affiliation(s)
- Yun Wu
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, P. R. China
| | - Yiming Qiao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Chenyu Yang
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, P. R. China
| | - Yueying Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Xizhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Chunhui Deng
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, P. R. China
| | - Qunyan Yao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
- Department of Gastroenterology and Hepatology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, P. R. China
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| |
Collapse
|
24
|
Quemerais C, Jean C, Brunel A, Decaup E, Labrousse G, Audureau H, Raffenne J, Belhabib I, Cros J, Perraud A, Dusetti N, Nicolle R, Mathonnet M, Pyronnet S, Martineau Y, Fanjul M, Bousquet C. Unveiling FKBP7 as an early endoplasmic reticulum sentinel in pancreatic stellate cell activation, collagen remodeling and tumor progression. Cancer Lett 2025; 614:217538. [PMID: 39924075 DOI: 10.1016/j.canlet.2025.217538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
In pancreatic ductal adenocarcinoma (PDAC), fibroblast activation leads to excessive secretion of extracellular matrix (ECM) and soluble factors that regulate tumor progression, prompting investigation into endoplasmic reticulum (ER)-resident proteins that may support this activation. We identified FKBP7, a peptidyl-prolyl isomerase in the ER, as overexpressed in PDAC stroma compared to cancer cells, and in patients with favorable prognosis. Analysis of single-cell RNA sequencing databases revealed FKBP7 expression in pancreatic stellate cells (PSCs) and cancer-associated fibroblasts (CAFs). When analyzed by immunohistochemistry on PDAC patient tissues, FKBP7 emerged as an early activation marker in the preneoplastic stroma, preceding αSMA expression, and responding to FAK- and TGFβ-induced stiffening and pro-fibrotic programs in PSCs. Functional analyses revealed that FKBP7 knockdown in PSCs enhanced contractility, Rho/FAK signaling, and secretion of pro-inflammatory cytokines as well as remodeling of type I collagen, promoting an activated phenotype and accelerating tumor growth in vivo. Conversely, FKBP7 expression supported a tumor-restraining (i.e. encapsulating) ECM characterized by type IV collagen. Mechanistically, FKBP7 interacts with BiP, and blocking this interaction instead leads to increased PSC secretion of type I collagen. Thus, FKBP7 serves as a novel PSC marker and ER regulator in a complex with BiP of the secretion of specific collagen subtypes, highlighting its potential to mediate ECM normalization and constrain PDAC tumorigenesis.
Collapse
Affiliation(s)
- Christophe Quemerais
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Christine Jean
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Alexia Brunel
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Emilie Decaup
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Guillaume Labrousse
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Hippolyte Audureau
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Jérôme Raffenne
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Ismahane Belhabib
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Jérôme Cros
- Department of Pathology, Beaujon-Bichat University Hospital - Paris Diderot University, Clichy, France
| | - Aurélie Perraud
- EA 3842 Laboratory, Medicine and Pharmacy Faculties, University of Limoges, France
| | - Nelson Dusetti
- Cancer Research Center of Marseille (CRCM), INSERM UMR-1068, CNRS UMR-7258, Marseille, France
| | - Remy Nicolle
- Center of Research on Inflammation (CRI), INSERM U1149, Paris, France
| | - Muriel Mathonnet
- EA 3842 Laboratory, Medicine and Pharmacy Faculties, University of Limoges, France
| | - Stéphane Pyronnet
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Yvan Martineau
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Marjorie Fanjul
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France
| | - Corinne Bousquet
- Cancer Research Center of Toulouse (CRCT), INSERM UMR-1037, CNRS UMR-5071, Team « Labellisée Ligue Contre le Cancer EL2021», University of Toulouse, France.
| |
Collapse
|
25
|
Sastre J, Pérez S, Sabater L, Rius-Pérez S. Redox signaling in the pancreas in health and disease. Physiol Rev 2025; 105:593-650. [PMID: 39324871 DOI: 10.1152/physrev.00044.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024] Open
Abstract
This review addresses oxidative stress and redox signaling in the pancreas under healthy physiological conditions as well as in acute pancreatitis, chronic pancreatitis, pancreatic cancer, and diabetes. Physiological redox homeodynamics is maintained mainly by NRF2/KEAP1, NF-κB, protein tyrosine phosphatases, peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α), and normal autophagy. Depletion of reduced glutathione (GSH) in the pancreas is a hallmark of acute pancreatitis and is initially accompanied by disulfide stress, which is characterized by protein cysteinylation without increased glutathione oxidation. A cross talk between oxidative stress, MAPKs, and NF-κB amplifies the inflammatory cascade, with PP2A and PGC1α as key redox regulatory nodes. In acute pancreatitis, nitration of cystathionine-β synthase causes blockade of the transsulfuration pathway leading to increased homocysteine levels, whereas p53 triggers necroptosis in the pancreas through downregulation of sulfiredoxin, PGC1α, and peroxiredoxin 3. Chronic pancreatitis exhibits oxidative distress mediated by NADPH oxidase 1 and/or CYP2E1, which promotes cell death, fibrosis, and inflammation. Oxidative stress cooperates with mutant KRAS to initiate and promote pancreatic adenocarcinoma. Mutant KRAS increases mitochondrial reactive oxygen species (ROS), which trigger acinar-to-ductal metaplasia and progression to pancreatic intraepithelial neoplasia (PanIN). ROS are maintained at a sufficient level to promote cell proliferation, while avoiding cell death or senescence through formation of NADPH and GSH and activation of NRF2, HIF-1/2α, and CREB. Redox signaling also plays a fundamental role in differentiation, proliferation, and insulin secretion of β-cells. However, ROS overproduction promotes β-cell dysfunction and apoptosis in type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Salvador Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Luis Sabater
- Liver, Biliary and Pancreatic Unit, Hospital Clínico, Department of Surgery, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Sergio Rius-Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
- Department of Cell Biology, Functional Biology and Physical Anthropology, Faculty of Biology, University of Valencia, Valencia, Spain
| |
Collapse
|
26
|
Duan H, Gao L, Asikaer A, Liu L, Huang K, Shen Y. Prognostic Model Construction of Disulfidptosis-Related Genes and Targeted Anticancer Drug Research in Pancreatic Cancer. Mol Biotechnol 2025; 67:1463-1482. [PMID: 38575817 DOI: 10.1007/s12033-024-01131-8] [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: 10/23/2023] [Accepted: 02/19/2024] [Indexed: 04/06/2024]
Abstract
Pancreatic cancer stands as one of the most lethal malignancies, characterized by delayed diagnosis, high mortality rates, limited treatment efficacy, and poor prognosis. Disulfidptosis, a recently unveiled modality of cell demise induced by disulfide stress, has emerged as a critical player intricately associated with the onset and progression of various cancer types. It has emerged as a promising candidate biomarker for cancer diagnosis, prognosis assessment, and treatment strategies. In this study, we have effectively established a prognostic risk model for pancreatic cancer by incorporating multiple differentially expressed long non-coding RNAs (DElncRNAs) closely linked to disulfide-driven cell death. Our investigation delved into the nuanced relationship between the DElncRNA-based predictive model for disulfide-driven cell death and the therapeutic responses to anticancer agents. Our findings illuminate that the high-risk subgroup exhibits heightened susceptibility to the small molecule compound AZD1208, positioning it as a prospective therapeutic agent for pancreatic cancer. Finally, we have elucidated the underlying mechanistic potential of AZD1208 in ameliorating pancreatic cancer through its targeted inhibition of the peroxisome proliferator-activated receptor-γ (PPARG) protein, employing an array of comprehensive analytical methods, including molecular docking and molecular dynamics (MD) simulations. This study explores disulfidptosis-related genes, paving the way for the development of targeted therapies for pancreatic cancer and emphasizing their significance in the field of oncology. Furthermore, through computational biology approaches, the drug AZD1208 was identified as a potential treatment targeting the PPARG protein for pancreatic cancer. This discovery opens new avenues for exploring targets and screening drugs for pancreatic cancer.
Collapse
Affiliation(s)
- Hongtao Duan
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Li Gao
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Aiminuer Asikaer
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Lingzhi Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Kuilong Huang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China
| | - Yan Shen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 405400, People's Republic of China.
| |
Collapse
|
27
|
Ma Z, Hua J, Wei M, Han L, Dong M, Xie W, Luo T, Meng Q, Wang W, Song Z, Shi S, Yu X, Xu J. The pancreatitis-cancer transformation-related factor, human rhomboid family-1, promotes pancreatic cancer progression through the SRC/YAP signaling pathway. Transl Oncol 2025; 54:102346. [PMID: 40056528 PMCID: PMC11930795 DOI: 10.1016/j.tranon.2025.102346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025] Open
Abstract
Pancreatic cancer is an aggressive malignancy characterized by rapid progression, unfavorable outcomes, and a low early detection rate. Elucidating the mechanisms underlying the onset and progression of pancreatic tumors is essential for early detection and for developing preventive measures. Even though human rhomboid family-1 (RHBDF) acts as an oncogene in various tumors, the role of RHBDF in pancreatic cancer progression remains unexplored. Here, publicly available datasets, including samples of chronic pancreatitis associated with pancreatic cancer from our center, were used for bioinformatics analyses, including differential expression, survival, and enrichment studies. The findings were validated by immunohistochemical staining and in vitro experiments. We found that RHBDF1 was significantly upregulated in tumor samples relative to adjacent non-tumor and pancreatitis tissues, and its expression increased in correlation with the progression of pancreatitis to cancer. Furthermore, RHBDF1 promoted the proliferation, migration, and invasion of pancreatic cancer cells, and in vivo studies demonstrated that RHBDF1 promoted pancreatic cancer progression, tissue fibrosis, and the formation of new blood vessels. RNA-sequencing and cell functional experiments indicated that RHBDF1 promotes the progression of pancreatic cancer through the SRC-YAP signaling pathway. In summary, the pancreatitis-cancer transformation-related factor, RHBDF1, promotes pancreatic cancer progression by activating the SRC-YAP signaling cascade, indicating that RHBDF1 could be a viable target for the diagnosis and treatment of early-stage pancreatic cancer.
Collapse
Affiliation(s)
- Zhilong Ma
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Jie Hua
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Miaoyan Wei
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Lin Han
- Central Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Mingwei Dong
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Wangcheng Xie
- Department of Hepatopancreatobiliary Surgery, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai 200434, China
| | - Tingyi Luo
- Department of Hepatopancreatobiliary Surgery, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai 200434, China
| | - Qingcai Meng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Zhenshun Song
- Department of Hepatopancreatobiliary Surgery, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai 200434, China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China.
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China.
| |
Collapse
|
28
|
Pienkowski T, Wawrzak-Pienkowska K, Tankiewicz-Kwedlo A, Ciborowski M, Kurek K, Pawlak D. Leveraging glycosylation for early detection and therapeutic target discovery in pancreatic cancer. Cell Death Dis 2025; 16:227. [PMID: 40164585 PMCID: PMC11958638 DOI: 10.1038/s41419-025-07517-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/17/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025]
Abstract
Pancreatic cancer (PC) remains one of the most lethal malignancies, primarily due to late-stage diagnosis, limited biomarker specificity, and aggressive metastatic potential. Recent glycoproteomic studies have illuminated the crucial role of glycosylation in PC progression, revealing altered glycosylation patterns that impact cell adhesion, immune evasion, and tumor invasiveness. Biomarkers such as CA19-9 remain the clinical standard, yet limitations in sensitivity and specificity, especially in early disease stages, necessitate the exploration of alternative markers. Emerging glycoproteins-such as mesothelin, thrombospondin-2, and glycan modifications like sialyl-Lewis x-offer diagnostic promise when combined with CA19-9 or used in profiling panels. Furthermore, therapeutic strategies targeting glycosylation processes, including sialylation, and fucosylation, have shown potential in curbing PC metastasis and enhancing immune response. Translational platforms, such as patient-derived xenografts and advanced in vitro models, are pivotal in validating these findings and assessing glycosylation potential therapeutic impact. Continued exploration of glycosylation-driven mechanisms and biomarker discovery in PC can significantly advance early detection and treatment efficacy, offering new hope in the management of this challenging disease.
Collapse
Affiliation(s)
- Tomasz Pienkowski
- Clinical Research Center, Medical University of Bialystok, Sklodowskiej MC 24A, Bialystok, Poland
- Department of Pharmacodynamics, Medical University of Bialystok, Bialystok, Poland
| | - Katarzyna Wawrzak-Pienkowska
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
- Department of Gastroenterology, Hepatology and Internal Diseases, Voivodeship Hospital in Bialystok, Bialystok, Poland
| | | | - Michal Ciborowski
- Clinical Research Center, Medical University of Bialystok, Sklodowskiej MC 24A, Bialystok, Poland
| | - Krzysztof Kurek
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
29
|
Bhattacharjee K, Sengupta A, Kumar R, Ghosh A. Identification of key hub genes in pancreatic ductal adenocarcinoma: an integrative bioinformatics study. FRONTIERS IN BIOINFORMATICS 2025; 5:1536783. [PMID: 40226632 PMCID: PMC11985535 DOI: 10.3389/fbinf.2025.1536783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/03/2025] [Indexed: 04/15/2025] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) poses a significant health threat characterized by poor clinical outcomes, largely attributable to late detection, chemotherapy resistance, and the absence of tailored therapies. Despite progress in surgical, radiation, and chemotherapy treatments, 80% of PDAC patients do not benefit optimally from systemic therapy, often due to asymptomatic presentation or disease regression upon diagnosis. The disease's progression is influenced by complex interactions involving immunological, genetic, and environmental factors, among others. However, the precise molecular mechanisms underlying PDAC remain incompletely understood. A major challenge in elucidating PDAC's origins lies in deciphering the genetic variations governing its network. PDAC exhibits heterogeneity, manifesting diverse genetic compositions, cellular attributes, and behaviors across patients and within tumors. This diversity complicates diagnosis, treatment strategies, and prognostication. Identification of "Differentially Expressed Genes" (DEGs) between PDAC and healthy controls is vital for addressing these challenges. These DEGs serve as the foundation for constructing the PDAC protein interaction network, with their network properties being assessed for further insights. Our analysis revealed five key hub genes (KHGs): EGF, SRC, SDC1, ICAM1 and CEACAM5. The KHGs were predominantly enriched in pathways such as: ErbB signaling pathway, Rap1 signaling pathway, etc. Acknowledging the therapeutic promise and biomarker importance of PDAC KHGs, we have also pinpointed approved medications for the identified key genes. Nevertheless, it is crucial to conduct experimental validation on KHGs to confirm their effectiveness within the PDAC context. Overall, this study identified potential key hub genes implicated in the progression of PDAC, offering significant guidance for personalized clinical decision-making and molecular-targeted therapy for PDAC patients.
Collapse
Affiliation(s)
| | - Avik Sengupta
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Rahul Kumar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Aryya Ghosh
- Department of Chemistry, Ashoka University, Sonipat, Haryana, India
| |
Collapse
|
30
|
Khabipov A, Miebach L, Lenz M, Kersting S, Bekeschus S. RAW264.7 Macrophages as a Polarization Model in the Context of Pancreatic Cancer and Chemokine Release. BIOLOGY 2025; 14:320. [PMID: 40282185 PMCID: PMC12024713 DOI: 10.3390/biology14040320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
The TME is a critical niche for determining the fate of cancer therapy. Tumor cells often polarize nontumor cells, including immune cells, in the TME to favor cancer growth. In pancreatic cancer, macrophages are associated with poor therapy outcomes and unfavorable survival, especially when rendered into M2 macrophages. The latter show features also found in so-called tumor-associated macrophages (TAM), which are described as protecting and propelling tumor growth. In this context, it has been understudied which pancreatic cancer chemokines contribute to macrophage polarization. To this end, we analyzed murine RAW264.7 macrophages and Panc02 and PDA6606 pancreatic cancer cells in mono- and coculture to identify release patterns of 13 chemokines. Artificial macrophage polarization confirmed prominent changes in surface receptor and chemokine secretion profiles. Strikingly, RAW264.7 cocultures with Panc02 or PDA6606 were congruent in showing elevated levels of CCL2, CCL5, CCL17, CCL20, CCL22, CXCL5, and CXCL10. Further underlining the suitability of our in vitro model, both pancreatic cancer cell lines showed similar modulation of the critical macrophage polarization markers arginase, CD206, and iNOS, as well as chemokine receptors CCR2 and CCR4. Collectively, we demonstrated that our model is suitable for testing the roles and functions of chemokines in macrophage polarization by pancreatic cancer cells.
Collapse
Affiliation(s)
- Aydar Khabipov
- Department of General, Visceral, Thoracic, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Lea Miebach
- Department of General, Visceral, Thoracic, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Maik Lenz
- Department of General, Visceral, Thoracic, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Stephan Kersting
- Department of General, Visceral, Thoracic, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Sander Bekeschus
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
- Department of Dermatology and Venerology, Rostock University Medical Center, Strempelstr. 13, 18057 Rostock, Germany
| |
Collapse
|
31
|
Chambers CR, Watakul S, Schofield P, Howell AE, Zhu J, Tran AMH, Kuepper N, Reed DA, Murphy KJ, Channon LM, Pereira BA, Tyma VM, Lee V, Trpceski M, Henry J, Melenec P, Abdulkhalek L, Nobis M, Metcalf XL, Ritchie S, Cadell A, Stoehr J, Magenau A, Chacon-Fajardo D, Chitty JL, O’Connell S, Zaratzian A, Tayao M, Da Silva A, Lyons RJ, Goldstein LD, Dale A, Rookyard A, Connolly A, Crossett B, Tran YTH, Kaltzis P, Vennin C, Dinevska M, Croucher DR, Samra J, Mittal A, Weatheritt RJ, Philp A, Del Monte-Nieto G, Zhang L, Enriquez RF, Cox TR, Shi YCC, Pinese M, Waddell N, Sim HW, Chtanova T, Wang Y, Joshua AM, Chantrill L, Evans TRJ, Gill AJ, Morton JP, Pajic M, Christ D, Herzog H, Timpson P, Herrmann D. Targeting the NPY/NPY1R signaling axis in mutant p53-dependent pancreatic cancer impairs metastasis. SCIENCE ADVANCES 2025; 11:eadq4416. [PMID: 40073121 PMCID: PMC11900870 DOI: 10.1126/sciadv.adq4416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 01/29/2025] [Indexed: 03/14/2025]
Abstract
Pancreatic cancer (PC) is a highly metastatic malignancy. More than 80% of patients with PC present with advanced-stage disease, preventing potentially curative surgery. The neuropeptide Y (NPY) system, best known for its role in controlling energy homeostasis, has also been shown to promote tumorigenesis in a range of cancer types, but its role in PC has yet to be explored. We show that expression of NPY and NPY1R are up-regulated in mouse PC models and human patients with PC. Moreover, using the genetically engineered, autochthonous KPR172HC mouse model of PC, we demonstrate that pancreas-specific and whole-body knockout of Npy1r significantly decreases metastasis to the liver. We identify that treatment with the NPY1R antagonist BIBO3304 significantly reduces KPR172HC migratory capacity on cell-derived matrices. Pharmacological NPY1R inhibition in an intrasplenic model of PC metastasis recapitulated the results of our genetic studies, with BIBO3304 significantly decreasing liver metastasis. Together, our results reveal that NPY/NPY1R signaling is a previously unidentified antimetastatic target in PC.
Collapse
Affiliation(s)
- Cecilia R. Chambers
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Supitchaya Watakul
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Peter Schofield
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Immune Biotherapies Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Anna E. Howell
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Jessie Zhu
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Alice M. H. Tran
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Nadia Kuepper
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Daniel A. Reed
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Kendelle J. Murphy
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Lily M. Channon
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Brooke A. Pereira
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Victoria M. Tyma
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Victoria Lee
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Michael Trpceski
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Jake Henry
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Immune Biotherapies Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Pauline Melenec
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Lea Abdulkhalek
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Max Nobis
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
| | - Xanthe L. Metcalf
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Shona Ritchie
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Antonia Cadell
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Janett Stoehr
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Astrid Magenau
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Diego Chacon-Fajardo
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Jessica L. Chitty
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Savannah O’Connell
- Immune Biotherapies Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Anaiis Zaratzian
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Michael Tayao
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Andrew Da Silva
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Ruth J. Lyons
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Leonard D. Goldstein
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Data Science Platform, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Ashleigh Dale
- Sydney Mass Spectrometry, University of Sydney, Sydney, New South Wales, Australia
| | - Alexander Rookyard
- Sydney Mass Spectrometry, University of Sydney, Sydney, New South Wales, Australia
| | - Angela Connolly
- Sydney Mass Spectrometry, University of Sydney, Sydney, New South Wales, Australia
| | - Ben Crossett
- Sydney Mass Spectrometry, University of Sydney, Sydney, New South Wales, Australia
| | - Yen T. H. Tran
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Peter Kaltzis
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Claire Vennin
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Marija Dinevska
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia
- Department of Surgery, University of Melbourne, Melbourne, Australia
| | | | | | - David R. Croucher
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Jaswinder Samra
- Royal North Shore Hospital, St Leonards, Sydney, New South Wales, Australia
| | - Anubhav Mittal
- Royal North Shore Hospital, St Leonards, Sydney, New South Wales, Australia
| | - Robert J. Weatheritt
- Immune Biotherapies Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Andrew Philp
- Centre for Healthy Ageing, Centenary Institute, Sydney, New South Wales, Australia
- School of Sport, Exercise and Rehabilitation Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Gonzalo Del Monte-Nieto
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Lei Zhang
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- St. Vincent’s Centre for Applied Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Ronaldo F. Enriquez
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Thomas R. Cox
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Yan-Chuan C. Shi
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Mark Pinese
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Hao-Wen Sim
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Tatyana Chtanova
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia
| | - Yingxiao Wang
- Department of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Anthony M. Joshua
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Lorraine Chantrill
- Department of Medical Oncology and Illawarra Shoalhaven Local Health District, Wollongong, New South Wales, Australia
| | - Thomas R. Jeffry Evans
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Anthony J. Gill
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- Royal North Shore Hospital, St Leonards, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Jennifer P. Morton
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Marina Pajic
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Translational Oncology Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
| | - Daniel Christ
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- Immune Biotherapies Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Herbert Herzog
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
- St. Vincent’s Centre for Applied Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - Paul Timpson
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| | - David Herrmann
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Kensington, Sydney, New South Wales, Australia
| |
Collapse
|
32
|
Tanaka J, Nakagawa T, Ono Y, Kamura Y, Ishida T, Kawabata H, Takahashi K, Sato H, Liss AS, Mizukami Y, Yokoi T. Highly multiplexed digital PCR assay for simultaneous quantification of variant allele frequencies and copy number alterations of KRAS and GNAS in pancreatic cancer precursors. Mol Oncol 2025. [PMID: 40077847 DOI: 10.1002/1878-0261.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 12/18/2024] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasms (IPMNs) are pancreatic ductal adenocarcinoma (PDAC) precursor lesions. Detecting these precursors and monitoring their progression are crucial for early PDAC diagnosis. Digital PCR (dPCR) is a highly sensitive nucleic acid quantification technique and offers a cost-effective option for patient follow-up. However, the clinical utility of conventional dPCR is restricted by multiplexing constraints, particularly due to the challenge of simultaneously quantifying multiple mutations and amplifications. In this study, we applied highly multiplexed dPCR and melting curve analysis to simultaneously measure single nucleotide mutations and amplifications of KRAS and GNAS. The developed 14-plex assay included both wild-type and mutant KRAS, a common driver gene in both PanIN and IPMN, and GNAS, which is specifically mutated in IPMN, along with RPP30, a reference gene for copy number alterations (CNAs). This multiplex dPCR method detected all target mutations with a limit of detection below 0.2% while quantifying CNAs. Additionally, the assay accurately quantified variant allele frequencies in liquid biopsy and tissue samples from both pancreatic neoplasm precursor and PDAC patients, indicating its potential for use in comprehensive patient follow-up.
Collapse
Affiliation(s)
- Junko Tanaka
- Center for Digital Services - Healthcare, Research & Development Group, Hitachi, Ltd., Tokyo, Japan
| | - Tatsuo Nakagawa
- Center for Digital Services - Healthcare, Research & Development Group, Hitachi, Ltd., Tokyo, Japan
| | - Yusuke Ono
- Institute of Biomedical Research, Sapporo Higashi Tokushukai Hospital, Japan
- Department of Advanced Genomic Community Healthcare, Asahikawa Medical University, Japan
| | - Yoshio Kamura
- Center for Digital Services - Healthcare, Research & Development Group, Hitachi, Ltd., Tokyo, Japan
| | - Takeshi Ishida
- Center for Digital Services - Healthcare, Research & Development Group, Hitachi, Ltd., Tokyo, Japan
| | - Hidemasa Kawabata
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Japan
| | - Kenji Takahashi
- Department of Advanced Genomic Community Healthcare, Asahikawa Medical University, Japan
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Japan
| | - Hiroki Sato
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Japan
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew S Liss
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yusuke Mizukami
- Institute of Biomedical Research, Sapporo Higashi Tokushukai Hospital, Japan
- Department of Advanced Genomic Community Healthcare, Asahikawa Medical University, Japan
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Japan
| | - Takahide Yokoi
- Center for Digital Services - Healthcare, Research & Development Group, Hitachi, Ltd., Tokyo, Japan
| |
Collapse
|
33
|
Wu HY, Tsou HH, Lu LS, Lee HL, Chiou JF, Ch'ang HJ. Role of Neoadjuvant Chemoradiation Therapy for Resectable and Borderline Resectable Pancreatic Adenocarcinoma-A Systematic Review and Meta-Analysis. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00175-0. [PMID: 40074045 DOI: 10.1016/j.ijrobp.2025.02.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/31/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Randomized trials and meta-analyses have indicated longer survival with neoadjuvant than with adjuvant therapy in patients with resectable or borderline resectable (R/BR) pancreatic adenocarcinoma. Despite the efficacy of chemotherapy, the role of radiation therapy as an adjuvant or neoadjuvant treatment for patients with R/BR pancreatic adenocarcinoma remains unclear. In this systematic review and meta-analysis, we compared the benefits of additional chemoradiation therapy (CRT) to neoadjuvant chemotherapy (NAC) with NAC alone for R/BR pancreatic adenocarcinoma. METHODS AND MATERIALS A systematic literature search was conducted on Embase, Web of Science, PubMed, Cochrane, and Google Scholar. Median overall survival (OS) was the primary endpoint. Secondary endpoints included disease-free survival (DFS), resection rate, and R0 resection rate. RESULTS This review and meta-analysis included 31 prospective studies, of which 9 were randomized trials. In these studies, 658 patients from 14 study arms received NAC alone and 912 patients from 19 study arms received both NAC and CRT (NAC-CRT). The pooled median OS was 25.55 months (95% CI, 21.59-30.24 months) for NAC alone and 17.55 months (95% CI, 16.47-18.70 months; P < .0001) for NAC-CRT. The pooled R0 resection rate was higher with NAC-CRT (83.43%) than with NAC (69.97%; P < .0001). No significant difference was observed in DFS or resection rate between the 2 groups. In patients who received 5 or more cycles of initial chemotherapy, NAC-CRT was associated with longer OS than NAC (23.30 vs 21.85 months; P = .856). CONCLUSIONS NAC provides significantly longer OS than NAC-CRT to R/BR pancreatic adenocarcinoma. NAC-CRT is associated with a significantly improved R0 resection rate. This positive local effect of CRT can be translated to extended survival when 5 cycles or more of NAC are prescribed.
Collapse
Affiliation(s)
- Hsiao-Yu Wu
- Institute of Public Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Hsiao-Hui Tsou
- Institute of Public Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Long-Sheng Lu
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hsin-Lun Lee
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Jeng Fong Chiou
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hui-Ju Ch'ang
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan; Department of Oncology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
34
|
Turlej E, Domaradzka A, Radzka J, Drulis-Fajdasz D, Kulbacka J, Gizak A. Cross-Talk Between Cancer and Its Cellular Environment-A Role in Cancer Progression. Cells 2025; 14:403. [PMID: 40136652 PMCID: PMC11940884 DOI: 10.3390/cells14060403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
The tumor microenvironment is a dynamic and complex three-dimensional network comprising the extracellular matrix and diverse non-cancerous cells, including fibroblasts, adipocytes, endothelial cells and various immune cells (lymphocytes T and B, NK cells, dendritic cells, monocytes/macrophages, myeloid-derived suppressor cells, and innate lymphoid cells). A constantly and rapidly growing number of studies highlight the critical role of these cells in shaping cancer survival, metastatic potential and therapy resistance. This review provides a synthesis of current knowledge on the modulating role of the cellular microenvironment in cancer progression and response to treatment.
Collapse
Affiliation(s)
- Eliza Turlej
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Aleksandra Domaradzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Justyna Radzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Dominika Drulis-Fajdasz
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Julita Kulbacka
- Departament of Molecular and Cellular Biology, Faculty of Pharmacy, Wrocław Medical University, Borowska 211A, 50-556 Wrocław, Poland;
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Agnieszka Gizak
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| |
Collapse
|
35
|
Phulara NR, Ishida CT, Espenshade PJ, Seneviratne HK. Gemcitabine Alters Phosphatidylcholine Metabolism in Mouse Pancreatic Tumors. J Proteome Res 2025; 24:1209-1218. [PMID: 39973059 DOI: 10.1021/acs.jproteome.4c00839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest diseases, despite advancements in elucidating tumor biology and developing novel therapeutics. Importantly, lipids, such as phospholipids, are crucial for the survival and proliferation of tumor cells. However, the impact of chemotherapeutic drugs on phospholipid metabolism in PDAC remains poorly understood. Gemcitabine (a nucleoside analogue) is a first-line drug in PDAC treatment, but its clinical effectiveness is limited by multiple factors. Herein, we employed matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) and proteomics approaches to investigate gemcitabine-induced lipid metabolism alterations in mouse pancreatic tumors following gemcitabine treatment (n = 3, control tumors; n = 3, gemcitabine-treated tumors). From MALDI MSI experiments, we observed elevated levels of several phosphatidylcholines (PCs), PC(30:0), PC(32:3), PC(34:2), PC(36:1), and PC(36:2), in gemcitabine-treated tumor tissues compared to the control. In addition, proteomics data revealed the differential abundance of several phospholipid-binding proteins in response to gemcitabine treatments. Furthermore, several endoplasmic reticulum stress-related proteins exhibited high expression in gemcitabine-treated tumor tissues. Altogether, our MALDI MSI and proteomics data provide important insights into alterations in PC metabolism in pancreatic tumors in response to gemcitabine treatment. Importantly, targeting the altered PC metabolism during gemcitabine therapy might help combat pancreatic cancer.
Collapse
Affiliation(s)
- Nav Raj Phulara
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| | - Chiaki Tsuge Ishida
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Peter John Espenshade
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Giovanis Institute for Translational Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Herana Kamal Seneviratne
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| |
Collapse
|
36
|
Cho YK, Kim S, Kim MJ, Lee WJ, Kim YJ, Jung CH. New users of sodium-glucose cotransporter 2 inhibitors are at low risk of incident pancreatic cancer: A nationwide population-based cohort study. DIABETES & METABOLISM 2025; 51:101605. [PMID: 39788347 DOI: 10.1016/j.diabet.2025.101605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
AIM We aimed to investigate whether sodium-glucose cotransporter-2 inhibitors (SGLT2is) are associated with a decreased risk of gastrointestinal (GI) cancers in patients with type 2 diabetes (T2D) compared to other glucose lowering medications (oGLMs). METHODS This active-comparator, new-user cohort study used the nationwide National Health Insurance Service database of the Republic of Korea from September 2014 to June 2020. From 79,423 new users of SGLT2is and 294,707 new users of oGLMs, we used a propensity score to match 59,954 from each of these two treatment groups. We calculated hazard ratios (HRs) and 95 % confidence intervals (CIs) for the incidence of GI cancers, encompassing stomach, colorectal, liver, and pancreatic cancers. RESULTS During the observation period, there were 814 and 916 GI cancers, and 794 and 1,140 deaths in the SGLT2is and oGLMs treatment groups, respectively. The use of SGLT2is was associated with a statistically significant reduction in the incidence of GI cancers, with an adjusted HR of 0.90 (95 % CI: 0.82 to 0.99). However, only the incidence of pancreatic cancer was significantly lower in SGLT2is users compared to non-users, with an adjusted HR of 0.72 (95 % CI: 0.55 - 0.95). In the entire cohort, the multivariable-adjusted HR for pancreatic cancer was 0.70 (95 % CI: 0.56 to 0.88). CONCLUSION For T2D patients, SGLT2i use was associated with a diminished pancreatic cancer risk compared to oGLMs. Future studies should ascertain the potential protective effect of SGLT2is against pancreatic cancer.
Collapse
Affiliation(s)
- Yun Kyung Cho
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Asan Diabetes Center, Asan Medical Center, Seoul, Republic of Korea
| | - Sehee Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Myung Jin Kim
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Asan Diabetes Center, Asan Medical Center, Seoul, Republic of Korea
| | - Woo Je Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Asan Diabetes Center, Asan Medical Center, Seoul, Republic of Korea
| | - Ye-Jee Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Chang Hee Jung
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Asan Diabetes Center, Asan Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
37
|
Lucas D, Sarkar T, Niemeyer CY, Harnoss JC, Schneider M, Strowitzki MJ, Harnoss JM. IRE1 is a promising therapeutic target in pancreatic cancer. Am J Physiol Cell Physiol 2025; 328:C806-C824. [PMID: 39819023 DOI: 10.1152/ajpcell.00551.2024] [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: 07/31/2024] [Revised: 08/13/2024] [Accepted: 01/14/2025] [Indexed: 01/19/2025]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Denise Lucas
- Department of General, Visceral, and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Tamal Sarkar
- Department of General, Visceral, Thoracic, and Transplant Surgery, University Hospital Giessen, Giessen, Germany
| | - Clara Y Niemeyer
- Department of General, Visceral, and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Julian C Harnoss
- Department of General, Visceral, and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral, Thoracic, and Transplant Surgery, University Hospital Giessen, Giessen, Germany
| | - Moritz J Strowitzki
- Department of General, Visceral, Thoracic, and Transplant Surgery, University Hospital Giessen, Giessen, Germany
| | - Jonathan M Harnoss
- Department of General, Visceral, and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
- Department of General, Visceral, Thoracic, and Transplant Surgery, University Hospital Giessen, Giessen, Germany
| |
Collapse
|
38
|
Zhong B, Du J, Liu F, Sun S. The Role of Yes-Associated Protein in Inflammatory Diseases and Cancer. MedComm (Beijing) 2025; 6:e70128. [PMID: 40066231 PMCID: PMC11892025 DOI: 10.1002/mco2.70128] [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: 04/17/2024] [Revised: 02/02/2025] [Accepted: 02/11/2025] [Indexed: 03/17/2025] Open
Abstract
Yes-associated protein (YAP) plays a central role in the Hippo pathway, primarily governing cell proliferation, differentiation, and apoptosis. Its significance extends to tumorigenesis and inflammatory conditions, impacting disease initiation and progression. Given the increasing relevance of YAP in inflammatory disorders and cancer, this study aims to elucidate its pathological regulatory functions in these contexts. Specifically, we aim to investigate the involvement and molecular mechanisms of YAP in various inflammatory diseases and cancers. We particularly focus on how YAP activation, whether through Hippo-dependent or independent pathways, triggers the release of inflammation and inflammatory mediators in respiratory, cardiovascular, and digestive inflammatory conditions. In cancer, YAP not only promotes tumor cell proliferation and differentiation but also modulates the tumor immune microenvironment, thereby fostering tumor metastasis and progression. Additionally, we provide an overview of current YAP-targeted therapies. By emphasizing YAP's role in inflammatory diseases and cancer, this study aims to enhance our understanding of the protein's pivotal involvement in disease processes, elucidate the intricate pathological mechanisms of related diseases, and contribute to future drug development strategies targeting YAP.
Collapse
Affiliation(s)
- Bing Zhong
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jintao Du
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Feng Liu
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Silu Sun
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesChinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and ManagementWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| |
Collapse
|
39
|
Tsilimigras DI, Woldesenbet S, Chatzipanagiotou OP, Iyer S, Pawlik TM. Long-term lorazepam use may be associated with worse long-term outcomes among patients with pancreatic adenocarcinoma. Surgery 2025; 179:108794. [PMID: 39304446 DOI: 10.1016/j.surg.2024.08.027] [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: 06/24/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Lorazepam recently has been reported to alter the tumor microenvironment of pancreatic adenocarcinoma in a murine model. We sought to evaluate whether the use of lorazepam was associated with worse outcomes among patients with pancreatic adenocarcinoma. METHODS Medicare beneficiaries diagnosed with stage I-IV pancreatic adenocarcinoma between 2013 and 2019 were identified from the Surveillance, Epidemiology and End Results-Medicare database. The association of lorazepam prescription relative to overall survival and recurrence-free survival was examined. RESULTS Among 2,810 patients with stage I-III and 10,181 patients with stage IV pancreatic adenocarcinoma, a total of 133 (4.7%) and 444 individuals (4.4%) had a lorazepam prescription before disease diagnosis, respectively. Although the overall lorazepam group had comparable 5-year overall survival (15.0% vs 14.2%, P = .20) and recurrence-free survival (12.7% vs 10.9%, P = .42) with the no-lorazepam group after pancreatic adenocarcinoma resection, individuals with long-term lorazepam prescription (>30 days) had worse 5-year overall survival (9.0% vs 21.0%, P = .02) and recurrence-free survival (6.4% vs 17.1%, P = .009) compared with short-term lorazepam users (≤30 days). Similarly, among patients with metastatic pancreatic adenocarcinoma, individuals with a long-term lorazepam prescription had worse 1-year overall survival (9.7% vs 15.9%, P = .02) compared with patients who had short-term lorazepam prescriptions. On multivariable analysis, long-term lorazepam prescription was independently associated with overall survival among patients with resectable (hazard ratio, 1.82; 95% confidence interval, 1.22-2.74) and metastatic pancreatic adenocarcinoma (hazard ratio, 1.24; 95% confidence interval, 1.02-1.51). CONCLUSION Long-term lorazepam prescription was associated with worse long-term outcomes among patients who underwent resection for pancreatic adenocarcinoma and patients with metastatic pancreatic adenocarcinoma. These data support the need for further large scale studies to confirm a potential harmful effect of lorazepam among patients with pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Diamantis I Tsilimigras
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH.
| | - Selamawit Woldesenbet
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH
| | - Odysseas P Chatzipanagiotou
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH
| | - Sidharth Iyer
- College of Medicine, The Ohio State University, Columbus, OH
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH.
| |
Collapse
|
40
|
Wang S, Pan T, Wang S, Zhang X, Peng L, Yang W. Impact of preoperative blood glucose levels on prognosis and postoperative complications in patients with pancreatic cancer. Oncol Lett 2025; 29:135. [PMID: 39839607 PMCID: PMC11747955 DOI: 10.3892/ol.2025.14880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025] Open
Abstract
The relationship between diabetes and pancreatic cancer is well documented; however, the effect of preoperative blood glucose levels on prognosis and postoperative complications is currently unclear. The present study aimed to investigate the effect of preoperative blood glucose levels on overall survival (OS) and postoperative complications in patients with pancreatic cancer. This retrospective study included 225 patients with pancreatic cancer treated at The Fourth Hospital of Hebei Medical University from January 2015 to December 2020. Patients were grouped based on preoperative blood glucose levels (normal, ≤6.11 mmol/l; high, >6.11 mmol/l). Data on demographics, clinical history, tumor characteristics, treatment and laboratory results were collected. High preoperative blood glucose levels were associated with reduced OS time [hazard ratio (HR), 1.68; 95% confidence interval (CI), 1.15-2.45; P=0.007] and increased postoperative complications (29.2 vs. 9.8%; χ2=13.658; P<0.001). Median OS time was significantly shorter in the high glucose group (14.2 vs. 20.5 months; HR, 1.96; 95% CI, 1.38-2.77; P<0.001). Elevated CA19-9 levels were also a predictor of poor OS (HR, 1.70; 95% CI, 1.06-2.74; P=0.029). High preoperative blood glucose and elevated CA19-9 levels were independent predictors of poor prognosis in patients with pancreatic cancer. This finding suggests that preoperative blood glucose levels have a greater impact on prognosis compared with a history of diabetes. Elevated preoperative blood glucose levels have poorer OS and a higher incidence of postoperative complications compared to those with lower preoperative glucose levels, underscoring the importance of preoperative glucose management. Effective preoperative blood glucose control may improve outcomes in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Shubin Wang
- Department of General Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Teng Pan
- Department of Oncology, Shijiazhuang First Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Shuya Wang
- Department of General Medicine, Dalian Medical University, Dalian, Liaoning 116000, P.R. China
| | - Xiaokun Zhang
- Department of General Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Li Peng
- Department of Hepatobiliary Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Wuhan Yang
- Department of Hepatobiliary Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
41
|
Shin SH, Lee YE, Yoon HN, Yuk CM, An JY, Seo M, Yoon S, Oh MS, Shin SC, Kim JH, Kim YJ, Kim JC, Kim SC, Jang M. An innovative strategy harnessing self-activating CAR-NK cells to mitigate TGF-β1-driven immune suppression. Biomaterials 2025; 314:122888. [PMID: 39423512 DOI: 10.1016/j.biomaterials.2024.122888] [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: 06/03/2024] [Revised: 09/19/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
The dysfunction of natural killer (NK) cells, mediated by transforming growth factor β1 (TGFβ1) within the tumor microenvironment, impedes antitumor therapy and contributes to poor clinical outcomes. Our study introduces self-activating chimeric antigen receptor (CAR)-NK cells that block TGFβ1 signaling by releasing a specifically designed peptide, P6, which targets mesothelin in pancreatic tumors. P6 originates from the interaction sites between TGFβ1 and TGFβ receptor 1 and effectively disrupts TGFβ1's inhibitory signaling in NK cells. Our analysis demonstrates that P6 treatment interrupts the SMAD2/3 pathway in NK cells, mitigating TGFβ1-mediated suppression of NK cell activity, thereby enhancing their metabolic function and cytotoxic response against pancreatic tumors. These CAR-NK cells exhibit potent antitumor capabilities, as evidenced in spheroid cultures with cancer-associated fibroblasts and in vivo mouse models. Our approach marks a substantial advancement in overcoming TGFβ1-mediated immune evasion, offering a promising avenue for revolutionizing cancer immunotherapy.
Collapse
Affiliation(s)
- Seung Hun Shin
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Republic of Korea; Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Young Eun Lee
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Han-Na Yoon
- Rare & Pediatric Cancer Branch, Division of Rare and Refractory Cancer, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Chae Min Yuk
- Center for Advanced Biomolecular Recognition, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Jun Yop An
- Corporate Research & Development Center, UCI Therapeutics, Seoul, Republic of Korea
| | - Minkoo Seo
- Corporate Research & Development Center, UCI Therapeutics, Seoul, Republic of Korea
| | - Sangwon Yoon
- Corporate Research & Development Center, UCI Therapeutics, Seoul, Republic of Korea
| | - Min-Suk Oh
- Corporate Research & Development Center, UCI Therapeutics, Seoul, Republic of Korea
| | - Sang Chul Shin
- Technological Convergence Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ji Hyung Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Yong Jun Kim
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea
| | - Jin-Chul Kim
- Natural Products Research Institute, Korea Institute of Science and Technology, Gangneung, Republic of Korea
| | - Song Cheol Kim
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Mihue Jang
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
42
|
Anand J, Droby G, Joseph S, Patel U, Zhang X, Klomp J, Der C, Purvis J, Wolff S, Bowser J, Vaziri C. TRIP13 protects pancreatic cancer cells against intrinsic and therapy-induced DNA replication stress. NAR Cancer 2025; 7:zcaf009. [PMID: 40115747 PMCID: PMC11923746 DOI: 10.1093/narcan/zcaf009] [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: 07/24/2024] [Revised: 01/25/2025] [Accepted: 03/11/2025] [Indexed: 03/23/2025] Open
Abstract
Oncogene activation in normal untransformed cells induces DNA replication stress and creates a dependency on DNA damage response (DDR) mechanisms for cell survival. Different oncogenic stimuli signal via distinct mechanisms in every cancer setting. The DDR is also pathologically reprogrammed and deployed in diverse ways in different cancers. Because mutant KRAS is the driver oncogene in 90% of pancreatic ductal adenocarcinomas (PDACs), here we have investigated DDR mechanisms by which KRAS-induced DNA replication stress is tolerated in normal human pancreatic epithelial cells [human pancreatic nestin-expressing (HPNE) cells]. Using a candidate screening approach, we identify TRIP13 as a KRASG12V-induced messenger RNA that is also expressed at high levels in PDAC relative to normal tissues. Using genetic and pharmacological tools, we show that TRIP13 is necessary to sustain ongoing DNA synthesis and viability specifically in KRASG12V-expressing cells. TRIP13 promotes survival of KRASG12V-expressing HPNE cells in a homologous recombination (HR)-dependent manner. KRASG12V-expressing HPNE cells lacking TRIP13 acquire hallmark HR deficiency phenotypes, including sensitivity to inhibitors of translesion synthesis and poly-ADP ribose polymerase. Established PDAC cell lines are also sensitized to intrinsic DNA damage and therapy-induced genotoxicity following TRIP13 depletion. Taken together, our results expose TRIP13 as an attractive new and therapeutically tractable vulnerability of KRAS-mutant PDAC.
Collapse
Affiliation(s)
- Jay R Anand
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Gaith N Droby
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Sayali Joseph
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Urvi Patel
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Xingyuan Zhang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Jeffrey A Klomp
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Channing J Der
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Jeremy E Purvis
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
- Computational Medicine Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Samuel C Wolff
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
- Computational Medicine Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Jessica L Bowser
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Cyrus Vaziri
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| |
Collapse
|
43
|
Świdnicka-Siergiejko A, Daniluk J, Miniewska K, Daniluk U, Guzińska-Ustymowicz K, Pryczynicz A, Dąbrowska M, Rusak M, Ciborowski M, Dąbrowski A. Inflammatory Stimuli and Fecal Microbiota Transplantation Accelerate Pancreatic Carcinogenesis in Transgenic Mice, Accompanied by Changes in the Microbiota Composition. Cells 2025; 14:361. [PMID: 40072088 PMCID: PMC11898920 DOI: 10.3390/cells14050361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 03/15/2025] Open
Abstract
An association between gut microbiota and the development of pancreatic ductal adenocarcinoma (PDAC) has been previously described. To better understand the bacterial microbiota changes accompanying PDAC promotion and progression stimulated by inflammation and fecal microbiota transplantation (FMT), we investigated stool and pancreatic microbiota by 16s RNA-based metagenomic analysis in mice with inducible acinar transgenic expressions of KrasG12D, and age- and sex-matched control mice that were exposed to inflammatory stimuli and fecal microbiota obtained from mice with PDAC. Time- and inflammatory-dependent stool and pancreatic bacterial composition alterations and stool alpha microbiota diversity reduction were observed only in mice with a Kras mutation that developed advanced pancreatic changes. Stool Actinobacteriota abundance and pancreatic Actinobacteriota and Bifidobacterium abundances increased. In contrast, stool abundance of Firmicutes, Verrucomicrobiota, Spirochaetota, Desulfobacterota, Butyricicoccus, Roseburia, Lachnospiraceae A2, Lachnospiraceae unclassified, and Oscillospiraceae unclassified decreased, and pancreatic detection of Alloprevotella and Oscillospiraceae uncultured was not observed. Furthermore, FMT accelerated tumorigenesis, gradually decreased the stool alpha diversity, and changed the pancreatic and stool microbial composition in mice with a Kras mutation. Specifically, the abundance of Actinobacteriota, Bifidobacterium and Faecalibaculum increased, while the abundance of genera such as Lachnospiraceace A2 and ASF356, Desulfovibrionaceace uncultured, and Roseburia has decreased. In conclusion, pancreatic carcinogenesis in the presence of an oncogenic Kras mutation stimulated by chronic inflammation and FMT dynamically changes the stool and pancreas microbiota. In particular, a decrease in stool microbiota diversity and abundance of bacteria known to be involved in short-fatty acids production were observed. PDAC mouse model can be used for further research on microbiota-PDAC interactions and towards more personalized and effective cancer therapies.
Collapse
Affiliation(s)
- Agnieszka Świdnicka-Siergiejko
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland; (J.D.); (A.D.)
| | - Jarosław Daniluk
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland; (J.D.); (A.D.)
| | - Katarzyna Miniewska
- Department of Medical Biochemistry, Medical University of Bialystok, 15-276 Bialystok, Poland;
| | - Urszula Daniluk
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition, Allergology and Pulmonology, Medical University of Bialystok, 15-276 Bialystok, Poland;
| | | | - Anna Pryczynicz
- Department of General Pathomorphology, Medical University of Bialystok, 15-276 Bialystok, Poland; (K.G.-U.); (A.P.)
| | - Milena Dąbrowska
- Department of Heamatological Diagnostics, Medical University of Bialystok, 15-276 Bialystok, Poland; (M.D.); (M.R.)
| | - Małgorzata Rusak
- Department of Heamatological Diagnostics, Medical University of Bialystok, 15-276 Bialystok, Poland; (M.D.); (M.R.)
| | - Michał Ciborowski
- Metabolomics and Proteomics Laboratory, Department of Medical Biochemistry, Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland;
| | - Andrzej Dąbrowski
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland; (J.D.); (A.D.)
| |
Collapse
|
44
|
Fang Z, Wu Z, Yu C, Xie Q, Zeng L, Chen R. EIF4E-mediated biogenesis of circPHF14 promotes the growth and metastasis of pancreatic ductal adenocarcinoma via Wnt/β-catenin pathway. Mol Cancer 2025; 24:56. [PMID: 40001070 PMCID: PMC11863466 DOI: 10.1186/s12943-025-02262-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND CircRNAs are critically involved in the development and progression of various cancers. However, their functions and mechanisms in pancreatic ductal adenocarcinoma (PDAC) remain largely unknown. METHODS CircPHF14 (hsa_circ_0079440) was identified through the analysis of RNA sequencing data from PDAC and normal adjacent tissues. The biological functions of circPHF14 were then evaluated using CCK8, EdU, transwell, colony formation, wound healing assays, as well as pancreatic orthotopic xenograft and liver metastasis models. The interaction mechanisms between circPHF14 and PABPC1, which enhance the stability of WNT7A mRNA, were investigated through RNA pull-down, mass spectrometry, RNA Immunoprecipitation (RIP), and actinomycin D assays. The role of EIF4E in promoting circPHF14 biogenesis was examined using RIP, and western blotting. RESULTS In this study, we observed a significant upregulation of circPHF14 in both clinical PDAC samples and cell lines. Functionally, circPHF14 enhanced PDAC proliferation and metastasis both in vitro and in vivo. Mechanistically, circPHF14 interacted with PABPC1 to stabilize WNT7A mRNA, thereby activating the Wnt/β-catenin pathway, which subsequently upregulated SNAI2 and initiated Epithelial-Mesenchymal Transition (EMT) in PDAC. Additionally, EIF4E was found to bind PHF14 pre-mRNA, facilitating circPHF14 biogenesis. Finally, we developed a lipid nanoparticle (LNP) formulation encapsulating sh-circPHF14 plasmids and confirmed its anti-tumor efficacy in a patient-derived xenograft (PDX) model. CONCLUSION EIF4E-mediated biogenesis of circPHF14 stabilizes WNT7A mRNA via interaction with PABPC1, which subsequently activates the Wnt/β-catenin pathway, promoting the growth and metastasis of PDAC. These findings indicate that circPHF14 holds promise as a biomarker and therapeutic target for PDAC.
Collapse
Affiliation(s)
- Zhou Fang
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhuo Wu
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chao Yu
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Qingyu Xie
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Liangtang Zeng
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Rufu Chen
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
45
|
Nasiri F, Safarzadeh Kozani P, Salem F, Mahboubi Kancha M, Dashti Shokoohi S, Safarzadeh Kozani P. Mechanisms of antigen-dependent resistance to chimeric antigen receptor (CAR)-T cell therapies. Cancer Cell Int 2025; 25:64. [PMID: 39994651 PMCID: PMC11849274 DOI: 10.1186/s12935-025-03697-y] [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: 10/08/2024] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
Cancer immunotherapy has reshaped the landscape of cancer treatment over the past decades. Genetic manipulation of T cells to express synthetic receptors, known as chimeric antigen receptors (CAR), has led to the creation of tremendous commercial and therapeutic success for the treatment of certain hematologic malignancies. However, since the engagement of CAR-T cells with their respective antigens is solely what triggers their cytotoxic reactions against target cells, the slightest changes to the availability and/or structure of the target antigen often result in the incapacitation of CAR-T cells to enforce tumoricidal responses. This results in the resistance of tumor cells to a particular CAR-T cell therapy that requires meticulous heeding to sustain remissions in cancer patients. In this review, we highlight the antigen-dependent resistance mechanisms by which tumor cells dodge being recognized and targeted by CAR-T cells. Moreover, since substituting the target antigen is the most potent strategy for overcoming antigen-dependent disease relapse, we tend to highlight the current status of some target antigens that might be considered suitable alternatives to the currently available antigens in various cancers. We also propose target antigens whose targeting might reduce the off-tumor adverse events of CAR-T cells in certain malignancies.
Collapse
Affiliation(s)
- Fatemeh Nasiri
- Department of Internal Medicine, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran
| | - Pouya Safarzadeh Kozani
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran.
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Faeze Salem
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maral Mahboubi Kancha
- Faculty of Engineering and Science, School of Science, University of Greenwich, Chatham Maritime, Chatham, Kent, ME4 4TB, UK
| | | | - Pooria Safarzadeh Kozani
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran.
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
46
|
Leiphrakpam PD, Chowdhury S, Zhang M, Bajaj V, Dhir M, Are C. Trends in the Global Incidence of Pancreatic Cancer and a Brief Review of its Histologic and Molecular Subtypes. J Gastrointest Cancer 2025; 56:71. [PMID: 39992560 DOI: 10.1007/s12029-025-01183-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2025] [Indexed: 02/25/2025]
Abstract
The global burden of pancreatic cancer has more than doubled in recent decades. It is now the sixth leading cause of cancer-related death worldwide, with an estimated 510,922 new cases and 467,409 deaths in 2022. The incidence of the disease continues to rise annually, with projections indicating a 95.4% increase in new cases by 2050, potentially reaching a total of 998,663 new cases globally. The overall five-year survival rate for pancreatic cancer is 10% worldwide, showing only a modest improvement compared to the past decade. The rising trends in the incidence rates are likely to continue as the global population ages and access to healthcare improves. The relatively low survival rate is primarily attributed to late-stage diagnoses and the lack of an effective screening method. Currently, population-based screening for asymptomatic individuals is not recommended, highlighting the importance of identifying and monitoring individuals at high risk for pancreatic cancer. Numerous studies have highlighted the differences in the molecular pathology of pancreatic cancer, underscoring the need for continued research to better understand these differences. The silent progression of the disease, poor prognosis, lack of screening options, and the necessity to improve our comprehension of its molecular characteristics emphasize the critical need for ongoing monitoring of disease trends at the population level. This review article analyses trends in the incidence of pancreatic cancer and its histological subtypes and provides an update on its molecular subtypes.
Collapse
Affiliation(s)
- Premila Devi Leiphrakpam
- Graduate Medical Education, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Surgical Oncology, Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sanjib Chowdhury
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michelle Zhang
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Varnica Bajaj
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mashaal Dhir
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Chandrakanth Are
- Graduate Medical Education, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
- Division of Surgical Oncology, Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
47
|
Mahdian SMA, Mahmoudi-Aznaveh A, Mousavi SM, Larijani B, Azizi Z, Javar HA. Plasma treatment can efficiently increase the attachment of pancreatic circulatory tumor cells to the surface. Discov Oncol 2025; 16:222. [PMID: 39982607 PMCID: PMC11845332 DOI: 10.1007/s12672-025-01988-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Pancreatic cancer ranks as the fourth most common cause of cancer-related fatalities globally, with a notably low 5-year relative survival rate. We need to immediately develop fast, dependable, and noninvasive diagnostic techniques that can accurately identify pancreatic cancer at an early stage. The research project created a straightforward but effective method for detecting and increasing the amount of tumor cells that could bind to polystyrene (PS) well plates. To significantly improve the adhesion of the pancreatic cancer cell line PANC-1 on PS well plates, a 5-min exposure to high-power oxygen plasma was implemented. This treatment caused a significant increase in surface energy and roughness. Surface characterization was assessed by utilizing an atomic force microscope and X-ray photoelectron spectroscopy. Water contact angle measurement is used to assess the level of wettability present on the treated surface. To determine how well the circulatory tumor cells (CTCs) model adheres to a plasma-treated surface (PTS), appropriate amounts of mCherry-labeled PANC-1 cells are mixed into a sample of blood cells to mimic clinical conditions. After applying plasma treatment, the experiment achieved a 96% success rate in binding at 2 h, specifically for the PANC-1 cell type. Moreover, the platform demonstrated a considerable ability to attach to cancerous cells compared to non-cancerous cells found in blood. To summarize, this study has shown that non-thermal plasma treatment could be a novel and efficient method for the better adhesion of pancreatic cancer cells, with the benefits of being cost-effective and quick. It is necessary for additional research to be conducted to confirm the clinical efficacy of the method.
Collapse
Affiliation(s)
- Seyed Mohammad Amin Mahdian
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Azam Mahmoudi-Aznaveh
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mojtaba Mousavi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Azizi
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hamid Akbari Javar
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
48
|
Britton E, Kobetic M, McNally E, Rudd S, Potter S, Hinchliffe R, Rees J. A systematic review of clinical outcome reporting for curative surgical treatment of patients with pancreatic adenocarcinoma. HPB (Oxford) 2025:S1365-182X(25)00067-X. [PMID: 40122766 DOI: 10.1016/j.hpb.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 12/06/2024] [Accepted: 02/15/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Pancreatic cancer is a leading cause of cancer-related death. Surgery (with systemic therapy) provides the only chance for long-term survival, but carries a high risk of morbidity and mortality. Robust evidence from meta-analyses, essential in informing decisions, is thwarted by inconsistencies between studies. This systematic review determines the nature and degree of heterogenous outcome reporting in research evaluating curative pancreatic cancer surgery. METHODS A literature search of Medline, Embase, Cochrane Central and clinicaltrials.gov from 2017 to 2023 for eligible randomised and prospective studies adhering to a PROSPERO registered protocol. RESULTS Included were 156 studies reporting a total of 2088 outcomes which deduplicated to 399 unique endpoints. No single outcome was reported in all studies. 45 % were not defined. Adverse events and delivery of care measures (typically technical aspects of surgery) accounted for 60 % and 32 % of outcomes respectively, compared to 6 % evaluating physical functioning post-surgery. CONCLUSION The vast number and diversity of outcomes in use demonstrates lack of discernment in choice and disparity over domains of importance. Further work is needed to embed uniform outcome definitions, harmonise data collection and refocus research on fewer outcomes of proven relevance. Developing consensus on these critical outcomes through a Core Outcome Set is recommended.
Collapse
Affiliation(s)
- Emily Britton
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol Royal Infirmary, Bristol, BS1 3NU, United Kingdom; University of Bristol Medical School, Learning & Research Centre, Southmead Hospital, Bristol, BS10 5FN, United Kingdom
| | - Matthew Kobetic
- University of Bristol Medical School, Learning & Research Centre, Southmead Hospital, Bristol, BS10 5FN, United Kingdom
| | - Eleanor McNally
- University of Bristol Medical School, Learning & Research Centre, Southmead Hospital, Bristol, BS10 5FN, United Kingdom
| | - Sarah Rudd
- North Bristol NHS Trust, Southmead Hospital, Bristol, BS10 5NB, United Kingdom
| | - Shelley Potter
- University of Bristol Medical School, Learning & Research Centre, Southmead Hospital, Bristol, BS10 5FN, United Kingdom; North Bristol NHS Trust, Southmead Hospital, Bristol, BS10 5NB, United Kingdom
| | - Robert Hinchliffe
- University of Bristol Medical School, Learning & Research Centre, Southmead Hospital, Bristol, BS10 5FN, United Kingdom; North Bristol NHS Trust, Southmead Hospital, Bristol, BS10 5NB, United Kingdom
| | - Jonathan Rees
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol Royal Infirmary, Bristol, BS1 3NU, United Kingdom; University of Bristol Medical School, Learning & Research Centre, Southmead Hospital, Bristol, BS10 5FN, United Kingdom.
| |
Collapse
|
49
|
Chen J, Zhang X, Zhang G, Zhu F, Liu W. Serum-derived exosomal miR-7977 combined with miR-451a as a potential biomarker for pancreatic ductal adenocarcinoma. BMC Cancer 2025; 25:295. [PMID: 39972247 PMCID: PMC11837301 DOI: 10.1186/s12885-025-13659-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/05/2025] [Indexed: 02/21/2025] Open
Abstract
OBJECTIVES To explore the potential of serum exosomal miRNAs as novel biomarkers for pancreatic ductal adenocarcinoma (PDAC). METHODS Serum exosomal miRNAs were screened and verified by microarray analysis and quantitative real-time PCR (qRT-PCR) in patients with PDAC and healthy controls. The correlation between the clinical characteristics of PDAC and candidate exosomal miRNAs was analyzed, and the diagnostic performance of the candidate biomarkers was evaluated. RESULTS Serum exosomal miR-7977 and miR-451a were significantly upregulated in PDAC patients compared with healthy controls, and the levels of miR-7977 and miR-451a in serum exosomes were closely associated with the clinical stage and metastasis of PDAC patients. The area under curve (AUC) values of serum exosomal miR-7977 and miR-451a for PDAC were 0.825 and 0.804 in the training set and 0.796 and 0.830 in the validation set, respectively. A biomarker panel consisting of these two miRNAs resulted in a diagnostic power with an AUC of 0.901 in the training set and 0.918 in the validation set. CONCLUSIONS Serum exosomal miR-7977 and miR-451a might be diagnostic biomarkers for PDAC. These two miRNAs, when combined, exhibit optimal diagnostic performance.
Collapse
Affiliation(s)
- Jia Chen
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Xue Zhang
- Department of Pathology, Affiliated Hospital of Chengdu University, Chengdu, 610081, China
| | - Guanyi Zhang
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Fan Zhu
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Disease Hospital, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, 650000, China.
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| | - Weiwei Liu
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
50
|
Jiang X, Zhu Y, Li J, Li W, Zheng W, Xu C, Zhang G. Laparoscopic radical antegrade modular pancreatosplenectomy vesus laparoscopic distal pancreatosplenectomy for left-sided pancreatic cancer: a systematic review and meta-analysis. Front Oncol 2025; 15:1510342. [PMID: 40027121 PMCID: PMC11867953 DOI: 10.3389/fonc.2025.1510342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/24/2025] [Indexed: 03/05/2025] Open
Abstract
Objective We aimed to compare the perioperative outcomes and postoperative complications of laparoscopic radical antegrade modular pancreatosplenectomy (L-RAMPS) versus laparoscopic distal pancreatosplenectomy (L-DPS) for left-sided pancreatic cancer through a meta-analysis. Methods A systematic review and meta-analysis, conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, were performed. Literature searches were conducted in PubMed, Web of Science, Cochrane Library, and Embase for studies published from their inception up to June 14th, 2024. Results A total of three retrospective studies involving 242 patients were included in this meta-analysis, with 116 patients in the L-RAMPS group and 126 in the L-DPS group. The meta-analysis results indicated that L-RAMPS was associated with the retrieval of more lymph nodes (MD: 3.06; 95% CI: 2.51 to 3.62, p < 0.00001) and longer operative time (MD: 20.05; 95% CI: 13.97 to 26.12, p < 0.00001) compared to L-DPS for left-sided pancreatic cancer patients. However, no significant differences were observed between the two groups in terms of R0 resection margins, the incidence of pancreatic fistula (Grade B and C), postpancreatectomy hemorrhage, or postoperative complications (Clavien-Dindo Grades II and III). Conclusions In patients with left-sided pancreatic cancer, L-RAMPS resulted in the retrieval of more lymph nodes, a longer operative time, and a similar incidence of postoperative complications compared to L-DPS. Larger sample sizes, extended follow-up periods, and well-conducted randomized controlled trials are needed to further validate these findings. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=558977, identifier CRD42024558977.
Collapse
Affiliation(s)
- Xutao Jiang
- Department of General Surgery, the Second Hospital of Dalian Medical University, Dalian, China
- Department of General Surgery, Dongxiang District People's Hospital, Fuzhou, China
| | - Yu Zhu
- Department of General Surgery, Dongxiang District People's Hospital, Fuzhou, China
| | - Jianwei Li
- Department of Intensive Care Medicine, Dongxiang District People's Hospital, Fuzhou, China
| | - Wei Li
- Department of General Surgery, Dongxiang District People's Hospital, Fuzhou, China
| | - Weizong Zheng
- Department of General Surgery, Dongxiang District People's Hospital, Fuzhou, China
| | - Caiming Xu
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Comprehensive Cancer Center, Monrovia, CA, United States
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guixin Zhang
- Department of General Surgery, the Second Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|