1
|
Zaki-Dizaji M, Taheri Z, Heiat M, Hushmandi K. Tumor-educated platelet, a potential liquid biopsy biosource in pancreatic cancer: A review. Pathol Res Pract 2025; 270:155986. [PMID: 40286788 DOI: 10.1016/j.prp.2025.155986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 04/13/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
Pancreatic cancer (PC) is a frequent and aggressive digestive system cancer with a very poor prognosis. The best chance for recovery lies in early surgical removal of the tumor. Unfortunately, because PC often develops without noticeable symptoms, diagnosis is frequently delayed. Limited treatment options, the metastasis potential of pancreatic cancer cells, and its generally poor prognosis mean that patients are often diagnosed late, significantly reducing the effectiveness of treatment. Consequently, there's a critical need for new biomarkers and technologies to improve early detection through screening. Recently, the liquid biopsy has developed as a powerful means for detecting and monitoring cancer at the molecular level. Its advantages include the ease and non-invasive nature of sample collection and its ability to reflect the dynamic changes within a tumor. Platelets, the second most numerous type of blood cell, offer a particularly promising source for liquid biopsy. It is known that cancer affects various aspects of platelets, including their number, size, activation state, and the proteins and RNA they contain. However, the full implications of these changes for cancer detection have not yet been fully integrated into routine clinical practice. Platelets have a unique ability to captivate nucleic acids and proteins from their surroundings, and they alter their transcriptome in response to external signals. This leads to the development of tumor-educated platelets (TEPs). Liquid biopsies that utilize TEP biomarkers hold considerable potential for screening, early detection, prognosis, guiding personalized treatment strategies, ongoing monitoring of the disease, and predicting recurrence. Encouraging results from preclinical studies have highlighted the potential of platelets as a novel liquid biopsy source for a wide range of cancers. This review will explore the potential of using platelets as a liquid biopsy method, specifically for pancreatic cancer.
Collapse
Affiliation(s)
- Majid Zaki-Dizaji
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Zahra Taheri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Lei J, Li X, Wang X, Xiao Y, Chi Y, Sun Q, Zhang H. Research on LCN2 interference to enhance the sensitivity of drug-resistant strains to gemcitabine. Xenobiotica 2025:1-9. [PMID: 40340561 DOI: 10.1080/00498254.2025.2501591] [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: 01/09/2025] [Revised: 04/17/2025] [Accepted: 04/25/2025] [Indexed: 05/10/2025]
Abstract
The aim of this study was to observe the sensitivity of the resistant strains to gemcitabine by interfering with the LCN2.An AsPC-1 gemcitabine-resistant cell line (GEM-R) was generated. Based on GEM-R, a lentivirus-infected shRNA-transfected LCN2 cell line was established. The proliferation of LCN2-regulated GEM-R cells was evaluated using the CCK-8 test and the mRNA expression of Ki-67. The apoptosis level of each drug-resistant strain was detected by flow cytometry. The expression of Bax, Bcl-2, Akt, E-cadherin and Vimentin were detected by western blotting.A gemcitabine-resistant strain of AsPC-1 was successfully induced and constructed as an shRNA LCN2 strain based on GEM-R. The interference of LCN2 expression enhanced the tumour inhibition and pro-apoptotic level of gemcitabine, increased the Bax/Bcl-2 value, and decreased p-Akt/Akt value. Meanwhile, the expression of E-cadherin was enhanced while the expression of Vimentin was decreased.This study confirmed that LCN2 affects gemcitabine sensitivity by participating in apoptosis and EMT processes, which may provide potential for overcoming gemcitabine resistance.
Collapse
Affiliation(s)
- Jianjun Lei
- Department of Laboratory Animal Center, General Hospital of Northern Theater Command, Shenyang, China
| | - Xuehua Li
- Department of Training Center for Clinical Skills and Medical staff, General Hospital of Northern Theater Command, Shenyang, China
| | - Xinpei Wang
- Department of Laboratory Animal Center, General Hospital of Northern Theater Command, Shenyang, China
| | - Yuwei Xiao
- Department of Laboratory Animal Center, General Hospital of Northern Theater Command, Shenyang, China
| | - Yang Chi
- Department of Laboratory Animal Center, General Hospital of Northern Theater Command, Shenyang, China
| | - Qian Sun
- Department of Laboratory Animal Center, General Hospital of Northern Theater Command, Shenyang, China
| | - He Zhang
- Department of Laboratory Animal Center, General Hospital of Northern Theater Command, Shenyang, China
- Department of Training Center for Clinical Skills and Medical staff, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
3
|
Kiss Z, Berki TL, Maráz A, Horváth Z, Nagy P, Fábián I, Kovács V, Rokszin G, Surján G, Barcza Z, Kenessey I, Wéber A, Wittmann I, Molnár GA, Szabó TG, Buga V, Karamousouli E, Darida M, Abonyi-Tóth Z, Bertókné Tamás R, Fürtős VD, Bogos K, Moldvay J, Gálffy G, Tamási L, Müller V, Krasznai ZT, Ostoros G, Pápai-Székely Z, Branyiczkiné Géczy G, Hilbert L, Polgár C, Vokó Z. Overall Survival of Hungarian Cancer Patients Diagnosed Between 2011 and 2019, Based on the Health Insurance Fund Database. Cancers (Basel) 2025; 17:1670. [PMID: 40427166 DOI: 10.3390/cancers17101670] [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: 04/07/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Assessing cancer survival trends is crucial for monitoring progress in cancer management and prevention. As part of the broader HUN-CANCER EPI study, this analysis examined overall survival (OS) in the Hungarian cancer population between 2011 and 2019. Methods: Using data extracted from the Hungarian National Health Insurance Fund (NHIF) database, short- and long-term OS were estimated for various cancer types according to age, sex, and diagnostic period using Kaplan-Meier analysis. The study also identified cancer types with significant early mortality following diagnosis. Results: From 2011 to 2019, a total of 528,808 patients were diagnosed with cancer. During the 2015-2019 diagnostic period, the lowest 5-year OS rates were observed for esophageal (7.0%), pancreatic (10.7%), liver (12.5%), gallbladder (13.9%), and lung cancer (18.4%). Conversely, tumor types with better OS included testicular cancer (91.6%), thyroid cancer (89.0%), Hodgkin's lymphoma (84.0%), melanoma (78.6%), and breast cancer (74.1%). A notable proportion of deaths occurred within 2 months of diagnosis for liver (33.2%), pancreatic (27.9%), and gallbladder cancer (29.0%). Significant early mortality within 6 months post-diagnosis was also noted for esophageal (51.3%), stomach (42.9%), and lung cancer (41.7%). Conclusions: The HUN-CANCER EPI study conducted between 2011 and 2019 provides valuable insights into cancer survival patterns in Hungary, emphasizing the importance of early detection and targeted interventions to improve patient outcomes.
Collapse
Affiliation(s)
- Zoltán Kiss
- MSD Pharma Hungary Ltd., 1095 Budapest, Hungary
- Second Department of Medicine and Nephrology-Diabetes Centre, University of Pécs Medical School, 7624 Pécs, Hungary
| | - Tamás László Berki
- Center for Health Technology Assessment, Semmelweis University, 1085 Budapest, Hungary
| | - Anikó Maráz
- Department of Oncotherapy, University of Szeged, 6720 Szeged, Hungary
| | - Zsolt Horváth
- Department of Oncology, Bács-Kiskun County Teaching Hospital, 6000 Kecskemét, Hungary
| | - Péter Nagy
- Department of Molecular Immunology and Toxicology, National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary
- Department of Anatomy and Histology, HUN-REN-UVMB Laboratory of Redox Biology Research Group, University of Veterinary Medicine, 1078 Budapest, Hungary
- Chemistry Institute, University of Debrecen, 4032 Debrecen, Hungary
| | - Ibolya Fábián
- RxTarget Ltd., 5000 Szolnok, Hungary
- Department of Biostatistics, University of Veterinary Medicine, 1078 Budapest, Hungary
| | | | | | - György Surján
- Department of Deputy Chief Medical Officer II., National Public Health Center, 1097 Budapest, Hungary
- Institute of Digital Health Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - Zsófia Barcza
- Syntesia Medical Communications Ltd., 1065 Budapest, Hungary
| | - István Kenessey
- Hungarian National Cancer Registry and National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, 1085 Budapest, Hungary
| | - András Wéber
- Hungarian National Cancer Registry and National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary
| | - István Wittmann
- Second Department of Medicine and Nephrology-Diabetes Centre, University of Pécs Medical School, 7624 Pécs, Hungary
| | - Gergő Attila Molnár
- Second Department of Medicine and Nephrology-Diabetes Centre, University of Pécs Medical School, 7624 Pécs, Hungary
| | | | | | | | | | - Zsolt Abonyi-Tóth
- RxTarget Ltd., 5000 Szolnok, Hungary
- Department of Biostatistics, University of Veterinary Medicine, 1078 Budapest, Hungary
| | - Renáta Bertókné Tamás
- Department of Deputy Chief Medical Officer II., National Public Health Center, 1097 Budapest, Hungary
| | - Viktória Diána Fürtős
- Department of Deputy Chief Medical Officer II., National Public Health Center, 1097 Budapest, Hungary
| | - Krisztina Bogos
- Directorate of Institution, National Korányi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Judit Moldvay
- 1st Department of Pulmonology, National Korányi Institute of Pulmonology, 1121 Budapest, Hungary
- Department of Pulmonology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Gabriella Gálffy
- Pulmonology Center of the Reformed Church in Hungary, Department of Pulmonology, 2045 Törökbálint, Hungary
| | - Lilla Tamási
- Department of Pulmonology, Semmelweis University, 1085 Budapest, Hungary
| | - Veronika Müller
- Department of Pulmonology, Semmelweis University, 1085 Budapest, Hungary
| | - Zoárd Tibor Krasznai
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Gyula Ostoros
- Directorate of Institution, National Korányi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Zsolt Pápai-Székely
- Fejér County Szent György, University Teaching Hospital, 8000 Székesfehérvár, Hungary
| | | | - Lászlóné Hilbert
- Demographic Statistics Department, Hungarian Central Statistical Office, 1024 Budapest, Hungary
| | - Csaba Polgár
- National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary
- Department of Oncology, Semmelweis University, 1085 Budapest, Hungary
| | - Zoltán Vokó
- Center for Health Technology Assessment, Semmelweis University, 1085 Budapest, Hungary
- Syreon Research Institute, 1145 Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
| |
Collapse
|
4
|
Mehtar A, Wechsler J, Desterke C, Giron-Michel J, Bouzidi A, Burlion A, Louache F, Kahia-Tani S, Uzan G, Naserian S. Optimizing Detection of Circulating Tumor Cells in Breast Cancer: Unveiling New Markers for Clinical Applications. Int J Mol Sci 2025; 26:4714. [PMID: 40429857 DOI: 10.3390/ijms26104714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/02/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Breast cancer (BC) is a heterogeneous disease with high metastasis potential, especially in the bones, liver, and lungs. Circulating tumor cells (CTCs), which emerge from active tumors, represent an early step toward metastasis and are associated with poor prognosis. CTCs of carcinoma origin are believed to express EpCAM and cytokeratins (CKs), common epithelial markers that are frequently used to identify them. However, in practice, the most aggressive CTCs lose the expression of those markers, leading to the partial loss of important information. Thus, finding some novel markers that identify CTCs regardless of their heterogeneity is crucial. A specific bioinformatics workflow integrating primary tumor and diverse BC cell lines transcriptomic expression analysis was developed and compared with single CTC transcriptomic analyses. We have identified a set of genes that are overexpressed in primary BC cells and are commonly upregulated among BC cell lines. Fifty of them were also found to be expressed in BC CTCs by single-cell transcriptomic analysis. Further in silico sorting narrowed this list to 12 genes. Using ScreenCell technology to isolate cancer cells spiked into normal blood, we tested the protein expression of all corresponding genes in vitro using the double immunocytochemistry method and validated MARCKSL1, SLC9A3R1, and RHOD as the most expressed markers. We then isolated the CTCs of 40 LN-invaded BC patients and 18 healthy donors using ScreenCell technology and showed that the combination of these three markers resulted in significantly better recognition of CTCs compared to EpCAM and CK conventional markers. Employing these novel markers, we found a clear distinction between blood samples from patients and healthy donors. In conclusion, through a specific bioinformatics workflow, in addition to in vitro and further clinical validations, we found three novel markers to precisely identify CTCs. These markers, when used together, enable a significantly more efficient identification of CTCs compared to conventional epithelial markers.
Collapse
Affiliation(s)
| | | | - Christophe Desterke
- Faculté de Médecine du Kremlin Bicêtre, Université Paris-Saclay, INSERM UMRS-1310, 94805 Villejuif, France
| | - Julien Giron-Michel
- Orsay-Vallée Campus, Paris-Saclay University, 91190 Gif-sur-Yvette, France
- Inserm UMR-S-MD 1197, Paul-Brousse Hospital, 94800 Villejuif, France
| | | | | | - Fawzia Louache
- Orsay-Vallée Campus, Paris-Saclay University, 91190 Gif-sur-Yvette, France
- Inserm UMR-S-MD 1197, Paul-Brousse Hospital, 94800 Villejuif, France
| | - Samira Kahia-Tani
- Kahia Laboratory, Oran 31000, Algeria
- Genethical, Oran 31000, Algeria
| | - Georges Uzan
- Orsay-Vallée Campus, Paris-Saclay University, 91190 Gif-sur-Yvette, France
- Inserm UMR-S-MD 1197, Paul-Brousse Hospital, 94800 Villejuif, France
| | | |
Collapse
|
5
|
Winterroth F, Wang J, Wink O, Carelsen B, Dahl J, Thakor AS. A Theoretical Approach in Applying High-Frequency Acoustic and Elasticity Microscopy to Assess Cells and Tissues. Annu Rev Biomed Eng 2025; 27:283-305. [PMID: 39971347 DOI: 10.1146/annurev-bioeng-112823-103134] [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
Medical ultrasound is a diagnostic imaging modality used for visualizing internal organs; the frequencies typically used are 2-10 MHz. Scanning acoustic microscopy (SAM) is a form of ultrasound where frequencies typically exceed 50 MHz. Increasing the acoustic frequency increases the specimen's spatial resolution but reduces the imaging depth. The advantages of using SAM over conventional light and electron microscopy include the ability to image cells and tissues without any preparation that could kill or alter them, providing a more accurate representation of the specimen. After scanning the specimen, acoustic signals are merged into an image on the basis of changes in the impedance mismatch between the immersion fluid and the specimens. The acoustic parameters determining the image quality are absorption and scattering. Surface scans can assess surface characteristics of the specimen. SAM is also capable of elastography, that is, studying elastic properties to discern differences between healthy and affected tissues. SAM has significant potential for detection/analysis in research and clinical studies.
Collapse
Affiliation(s)
| | - Jing Wang
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Onno Wink
- Philips Research, Eindhoven, The Netherlands;
| | | | - Jeremy Dahl
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, California, USA
| |
Collapse
|
6
|
Tan T, Ma L, Guo Y, Chen T, Meng L, Luo K, Zhou P, Cai M, Ji M, Hu H. Intelligent Diagnosis of Pancreatic Biopsy From Endoscopic Ultrasound-Guided Fine-Needle Aspiration Via Stimulated Raman Histopathology. J Transl Med 2025; 105:104182. [PMID: 40288652 DOI: 10.1016/j.labinv.2025.104182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 04/06/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
Endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA) has become one of the most important preoperative diagnostic methods for pancreatic tumors, but it often faces challenges of redundant sampling from patients and complex tissue processing that hinders timely diagnosis. Intraoperative rapid on-site evaluation is an auxiliary diagnostic technique that helps assess sample quality in real time, but it heavily depends on pathologists and involves subjectivity and complex procedures. Here, we developed a rapid and label-free approach for intraoperative histology on EUS-FNA specimen via deep learning-based stimulated Raman scattering microscopy, aimed at replacing rapid on-site evaluation and providing a more efficient and objective diagnostic approach. Fresh pancreatic EUS-FNA tissues were imaged with stimulated Raman scattering and compared with hematoxylin and eosin staining to identify key histologic features. Using images from 76 patients, a convolutional neural network model was established to identify benign, malignant, and nondiagnostic areas, achieving a validation accuracy >96% on an external test set of 33 cases. Furthermore, gradient-weighted class activation mapping was able to highlight histologic profiles within individual biopsy. Our approach has potential application in efficient intraoperative assessment of pancreatic biopsy through EUS-FNA.
Collapse
Affiliation(s)
- Tao Tan
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Collaborative Innovation Center of Endoscopy, Zhongshan Hospital, Fudan University, Shanghai, China; School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Liyang Ma
- State Key Laboratory of Surface Physics and Department of Physics, Fudan University, Shanghai, China; Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Yuheng Guo
- State Key Laboratory of Surface Physics and Department of Physics, Fudan University, Shanghai, China; Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Tianyin Chen
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Collaborative Innovation Center of Endoscopy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lili Meng
- Pathology Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kuan Luo
- State Key Laboratory of Surface Physics and Department of Physics, Fudan University, Shanghai, China; Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Pinghong Zhou
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Collaborative Innovation Center of Endoscopy, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Mingyan Cai
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Collaborative Innovation Center of Endoscopy, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Minbiao Ji
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Collaborative Innovation Center of Endoscopy, Zhongshan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Surface Physics and Department of Physics, Fudan University, Shanghai, China; Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Academy for Engineering and Technology, Fudan University, Shanghai, China.
| | - Hao Hu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Collaborative Innovation Center of Endoscopy, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Noversa de Sousa R, Carqueja I, Sá Lima A, Guimarães F. Pancreatic cancer unveiled by multisystemic manifestations. BMJ Case Rep 2025; 18:e264969. [PMID: 40280580 DOI: 10.1136/bcr-2025-264969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025] Open
Abstract
Pancreatic cancer is characterised by its aggressive progression, late-stage diagnosis and poor prognosis. This case report illustrates the intricate interplay between pancreatic adenocarcinoma, disseminated intravascular coagulation with diffuse alveolar haemorrhage and bone metastasis in an elderly woman. Presenting with asthenia, significant hip pain and haemoptysis, her work-up revealed multiple supradiaphragmatic adenopathies and infradiaphragmatic adenopathies, pancreatic parenchymal heterogeneity and lytic bone lesions. Despite extensive diagnostic efforts, a diagnosis of adenocarcinoma of probable pancreatobiliary origin was established only at an advanced stage, leading to a fatal outcome within a week. The patient experienced rapid clinical deterioration, characterised by refractory respiratory failure, haematological dysfunction and circulatory shock, highlighting the aggressive course of pancreatic cancer and its complications. This case underscores the diagnostic challenges and the critical need for early recognition and tailored strategies in managing complex presentations of pancreatic cancer, especially with rare and severe systemic manifestations.
Collapse
Affiliation(s)
- Rita Noversa de Sousa
- Internal Medicine, Pedro Hispano Hospital, Matosinhos Local Health Unit, Matosinhos, Portugal
| | - Inês Carqueja
- Intensive Care Unit, Pedro Hispano Hospital, Matosinhos Local Health Unit, Matosinhos, Portugal
| | - Andreia Sá Lima
- Internal Medicine, Pedro Hispano Hospital, Matosinhos Local Health Unit, Matosinhos, Portugal
| | - Filipa Guimarães
- Intensive Care Unit, Pedro Hispano Hospital, Matosinhos Local Health Unit, Matosinhos, Portugal
| |
Collapse
|
8
|
Shenoy A, Yousif A, Hussain MD. Recent Advances and Challenges in the Treatment of Advanced Pancreatic Cancer: An Update on Completed and Ongoing Clinical Trials. Cancers (Basel) 2025; 17:1319. [PMID: 40282495 PMCID: PMC12025738 DOI: 10.3390/cancers17081319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/29/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025] Open
Abstract
Pancreatic cancer is a deadly disease with a low survival rate, particularly in its advanced stages. Advanced pancreatic cancer remains a major clinical challenge due to limited treatment options. Surgical resection may not always be feasible, and traditional chemotherapy often shows restricted effectiveness. As a result, researchers are exploring a multifaceted therapeutic approach targeting the genetic and molecular drivers of the disease. A combination of molecular profiling and targeted therapies are being investigated to improve outcomes and address the shortcomings of traditional treatments. The focus of this review is to provide a summary of current and completed clinical trials for the treatment of advanced pancreatic cancer. This includes adagrasib (a KRAS inhibitor), olaparib (a PARP inhibitor for BRCA mutations), APG-1387 (an IAP antagonist), minnelide (an anti-stromal agent), arimastat (an MMP inhibitor), MK-0646 (an IGF1R inhibitor), sirolimus (an mTOR inhibitor), and metabolic inhibitors. These agents are being evaluated both as standalone treatments and in combination with standard therapy. Furthermore, we have summarized novel approaches such as cancer vaccines and ablation techniques as emerging strategies in the treatment of advanced pancreatic cancer. We have also examined the challenges in treating advanced pancreatic cancer and the factors contributing to therapeutic failure, which may offer valuable insights for developing more effective treatment strategies and innovative drug designs.
Collapse
Affiliation(s)
- Abhinav Shenoy
- College of Engineering, Texas A&M University, College Station, TX 77843, USA;
| | - Amar Yousif
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA;
| | - Muhammad Delwar Hussain
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA;
| |
Collapse
|
9
|
Huhulea EN, Huang L, Eng S, Sumawi B, Huang A, Aifuwa E, Hirani R, Tiwari RK, Etienne M. Artificial Intelligence Advancements in Oncology: A Review of Current Trends and Future Directions. Biomedicines 2025; 13:951. [PMID: 40299653 PMCID: PMC12025054 DOI: 10.3390/biomedicines13040951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 05/01/2025] Open
Abstract
Cancer remains one of the leading causes of mortality worldwide, driving the need for innovative approaches in research and treatment. Artificial intelligence (AI) has emerged as a powerful tool in oncology, with the potential to revolutionize cancer diagnosis, treatment, and management. This paper reviews recent advancements in AI applications within cancer research, focusing on early detection through computer-aided diagnosis, personalized treatment strategies, and drug discovery. We survey AI-enhanced diagnostic applications and explore AI techniques such as deep learning, as well as the integration of AI with nanomedicine and immunotherapy for cancer care. Comparative analyses of AI-based models versus traditional diagnostic methods are presented, highlighting AI's superior potential. Additionally, we discuss the importance of integrating social determinants of health to optimize cancer care. Despite these advancements, challenges such as data quality, algorithmic biases, and clinical validation remain, limiting widespread adoption. The review concludes with a discussion of the future directions of AI in oncology, emphasizing its potential to reshape cancer care by enhancing diagnosis, personalizing treatments and targeted therapies, and ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Ellen N. Huhulea
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
| | - Lillian Huang
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
| | - Shirley Eng
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
| | - Bushra Sumawi
- Barshop Institute, The University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Audrey Huang
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
| | - Esewi Aifuwa
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
| | - Rahim Hirani
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
- Graduate School of Biomedical Sciences, New York Medical College, Valhalla, NY 10595, USA
| | - Raj K. Tiwari
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
- Graduate School of Biomedical Sciences, New York Medical College, Valhalla, NY 10595, USA
| | - Mill Etienne
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA (R.H.)
- Department of Neurology, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
10
|
Li J, Wang A, Guo H, Zheng W, Chen R, Miao C, Zheng D, Peng J, Wang J, Chen Z. Exosomes: innovative biomarkers leading the charge in non-invasive cancer diagnostics. Theranostics 2025; 15:5277-5311. [PMID: 40303340 PMCID: PMC12036879 DOI: 10.7150/thno.113650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 04/06/2025] [Indexed: 05/02/2025] Open
Abstract
Exosomes, nanoscale extracellular vesicles secreted by diverse cell types, have emerged as promising biomarkers for non-invasive tumor diagnostics, offering significant advantages over traditional methods. These vesicles, typically ranging from 30 to 150 nanometers in size, carry a diverse cargo of proteins, lipids, RNA, and microRNAs, which reflect the molecular alterations occurring within their parent cells. Notably, exosomes can be isolated from easily accessible biofluids such as blood, urine, and saliva, making them ideal candidates for liquid biopsy applications. This review explores the transformative potential of exosome-based biomarkers in the early detection and monitoring of cancers across diverse organ systems, including respiratory, digestive, hematological, neurological, endocrine malignancies and so on. Special emphasis is placed on their application in clinical trials, where exosome-based diagnostics have demonstrated promising results in detecting tumors at early stages and monitoring treatment responses, offering a less invasive and more accessible alternative to traditional biopsies. While recent advancements in exosome isolation and characterization technologies have significantly improved the sensitivity and specificity of these diagnostics, challenges such as biological heterogeneity, lack of standardization, and regulatory hurdles remain. Nevertheless, exosome-based diagnostics hold the promise of providing real-time, dynamic insights into tumor progression, enhancing personalized medicine. The integration of exosomes into clinical practice could revolutionize cancer diagnostics and therapy, improving patient outcomes. Further research and large-scale clinical validation are essential to fully realize the clinical potential of exosome-based biomarker applications in routine clinical settings.
Collapse
Affiliation(s)
- Jiale Li
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China, 570208
| | - Ailin Wang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China, 211198
| | - Haijun Guo
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, Hunan, China, 412000
| | - Wei Zheng
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, Hunan, China, 412000
| | - Rui Chen
- Department of Neurosurgery, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan, China, 533000
| | - Changfeng Miao
- Department of Neurosurgery Second Branche, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China, 410005
| | - Dandan Zheng
- Department of Radiation Oncology, The First Affiliated Hospital Zhejiang University, Hangzhou, China, 310009
| | - Jun Peng
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China, 570208
| | - Jiachong Wang
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China, 570208
| | - Zigui Chen
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China, 570208
| |
Collapse
|
11
|
Jeong HJ, Heo D, Lim SY, Kim HS, Chae H, Yoon SJ, Shin SH, Han IW, Heo JS, Min JH, Kim H. Impact of portal/superior mesenteric vein abutment angle on prognosis in pancreatic cancer: a single-center retrospective cohort study. Ann Surg Treat Res 2025; 108:231-239. [PMID: 40226170 PMCID: PMC11982446 DOI: 10.4174/astr.2025.108.4.231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/09/2024] [Accepted: 01/08/2025] [Indexed: 04/15/2025] Open
Abstract
Purpose Pancreatic cancer has a poor prognosis; however, the implementation of neoadjuvant treatment enables borderline resectable cases to undergo curative resection and improves the overall survival rate. Attempts have been made to expand the eligibility criteria for neoadjuvant treatment, even in resectable cases. Some studies have suggested a correlation between vein abutment and poor prognosis or that the abutment angle may affect prognosis. This study investigated the anatomical factors affecting the vessel abutment angle and its prognostic value in pancreatic cancer. Methods Patients with pancreatic ductal adenocarcinoma who underwent surgery between 2012 and 2017 were included in this study. Patients who underwent neoadjuvant treatment were excluded. Data from only the intent-to-treat pancreaticoduodenectomy group were included in the analysis. Clinicopathological characteristics; preoperative factors such as CA 19-9, preoperative biliary drainage, American Society of Anesthesiologists physical status classification, portal vein/superior mesenteric vein contact angle measured via CT scan; and intraoperative factors were collected for analysis. Results A total of 365 patients were included in this study, and the abutment group included 92 patients (25.2%). The abutment and no-contact groups did not show any significant differences in terms of the overall survival or disease-free survival rate. Among the abutment groups, patients with less than 90° and 90°-180° did not show any significant differences. In the multivariate analysis, the only preoperative factor that had a prognostic effect was CA 19-9, a biological factor. Conclusion When there is no vessel invasion in the abutment group, upfront surgery should be considered because the angle does not affect the overall prognosis.
Collapse
Affiliation(s)
- Hye Jeong Jeong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Surgery, Daejeon Eulji Medical Center, Eulji University School of Medicine, Daejeon, Korea
| | - DanHui Heo
- Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Soo Yeun Lim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Hyeong Seok Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hochang Chae
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - So Jeong Yoon
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang Hyun Shin
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - In Woong Han
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin Seok Heo
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Hye Min
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hongbeom Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
12
|
Wu Y, Zhang C, Huang J, Chen Q, Zhang Y, Liu F, Xu D, Jiang K, Shi R, Chen M, Yuan H. Integrated analysis of scRNA-seq and bulk RNA-seq data identifies BHLHE40 as a key gene in pancreatic cancer progression and gemcitabine resistance. Semin Oncol 2025; 52:152338. [PMID: 40250076 DOI: 10.1016/j.seminoncol.2025.152338] [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: 02/17/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 04/20/2025]
Abstract
OBJECTIVE Pancreatic cancer is characterized by its high mortality rate and short survival periods, and novel therapeutic targets and tailor personalized strategies are urgently needed. In this study, we aim to investigate the molecular mechanisms underlying pancreatic ductal adenocarcinoma (PDAC) progression and chemoresistance, with a focus on identifying novel therapeutic targets. METHODS Multiomics approaches were integrated to identify novel actionable targets for PDAC. Public datasets such as TCGA and GEO were utilized to investigate the relationship between gene expression and clinical outcomes. Functional enrichment, cell-cell communication, and metabolic pathway analyses were performed to reveal PDAC heterogeneity and therapeutic resistance mechanisms. RESULTS BHLHE40 was identified as a hub gene linked to high-CNV PDAC cells, Gemcitabine resistance, and poor prognosis in PDAC. High BHLHE40 expression is significantly correlated with immunosuppressive tumor microenvironment (TME) features such as reduced CD8+ T infiltration, TCR richness, and lower tumor mutational burden (TMB). ChIP-seq data analysis confirmed BHLHE40 could directly bind to the SAT1 promoter, establishing a transcriptional axis promoting chemoresistance. Single-cell RNA-seq analysis further revealed that the BHLHE40+/SAT1+ subpopulation cells are resistant to Gemcitabine in PDAC. CONCLUSIONS BHLHE40 is significantly correlated with PDAC malignancy and chemoresistance via SAT1 regulation and immune evasion. Targeting BHLHE40 may sensitize PDACs to Gemcitabine and facilitate personalized treatment for BHLHE40+ PDAC patients.
Collapse
Affiliation(s)
- Yang Wu
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chun Zhang
- Department of Gastroenterology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiacheng Huang
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qun Chen
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yufeng Zhang
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fengyuan Liu
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dong Xu
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kuirong Jiang
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mengxing Chen
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Yuan
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
13
|
Guo K, Li S, Wu X, Xiong H. Nanomedicine in the Diagnosis and Treatment of Pancreatic Cancer. Pharmaceutics 2025; 17:449. [PMID: 40284444 PMCID: PMC12030228 DOI: 10.3390/pharmaceutics17040449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with increasing incidence and mortality rates, highlighting the urgent need for early diagnosis and treatment. However, early diagnosis of PDAC is extremely challenging due to the atypical early symptoms or the absence of noticeable symptoms. As a result, many patients are diagnosed with local metastasis, and even patients who are eligible for surgical resection have a high postoperative recurrence rate. Consequently, chemotherapy remains the primary treatment for PDAC. However, the unique biological characteristics of PDAC not only promote tumor progression and metastasis but also often lead to chemoresistance, a significant barrier to successful treatment. Recently, nanomaterials have garnered significant attention as promising materials for diagnosing and treating PDAC, showing great potential in cancer therapy, imaging, and drug delivery. Novel targeted nanomedicines, which encapsulate chemotherapy drugs and gene therapy products, offer significant advantages in overcoming resistance. These nanomedicines not only provide innovative solutions to the limitations of conventional chemotherapy but also improve the selectivity for cancer cells to enhance therapeutic outcomes. Current research is focused on the development of advanced nanomedicines, such as liposomes, nanotubes, and polymer-lipid hybrid systems, aimed at making chemotherapy more effective and longer lasting. This review provides a detailed overview of various nanomedicines utilized in the diagnosis and treatment of PDAC and outlines future directions for their development and key breakthroughs.
Collapse
Affiliation(s)
| | | | - Xinyu Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (K.G.); (S.L.)
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (K.G.); (S.L.)
| |
Collapse
|
14
|
Zhang J, Sun W, Wu W, Qin Z, Wei B, Li T. METTL3-dependent m6A methylation of circCEACAM5 fuels pancreatic cancer progression through DKC1 activation. Cell Mol Life Sci 2025; 82:132. [PMID: 40146281 PMCID: PMC11950576 DOI: 10.1007/s00018-025-05653-5] [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: 11/14/2024] [Revised: 02/20/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Pancreatic cancer is highly lethal and has a poor prognosis. Research has highlighted the role of circular RNAs and m6A methylation in cancer progression. METTL3, a key m6A methyltransferase, is linked to various cancers, but its interaction with circular RNAs in pancreatic cancer is unclear. This study examined the role of circCEACAM5 in pancreatic cancer, particularly its regulation by METTL3-mediated m6A methylation and interaction with effectors such as DKC1. METHODS circCEACAM5 expression in pancreatic cancer tissues and cell lines was evaluated via RT‒qPCR. Its characteristics were validated through Sanger sequencing, stability assays, and FISH. Functional assays (CCK-8, EdU, Transwell, and flow cytometry) were conducted in AsPC-1 cells, and in vivo tumor models were established. m6A modification was analyzed via bioinformatics tools and m6A-specific immunoprecipitation, while RNA pull-down assays were used to examine the interaction of circCEACAM5 with METTL3 and DKC1. RESULTS circCEACAM5 was significantly upregulated in pancreatic cancer and correlated with poor clinical outcomes. CircCEACAM5 promoted cell proliferation, invasion, and migration while inhibiting apoptosis both in vitro and in vivo. METTL3-mediated m6A methylation of circCEACAM5 was confirmed, and METTL3 knockdown reversed the effects of circCEACAM5 silencing on the malignant behavior of pancreatic cancer cells. circCEACAM5 interacted with DKC1, and DKC1 overexpression reversed the effects of circCEACAM5 knockdown on the malignant behavior of pancreatic cancer cells. CONCLUSION METTL3-mediated m6A methylation of circCEACAM5 drives pancreatic cancer progression by increasing DKC1 expression, suggesting potential new therapeutic targets for this aggressive malignancy.
Collapse
Affiliation(s)
- Jie Zhang
- School of Biology and Food Engineering, Changshu Institute of Technology, Suzhou, 215500, People's Republic of China
| | - Wenxue Sun
- Jining First People's Hospital, Jining Medical University, Jining, 272000, People's Republic of China
| | - Wenda Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Zihui Qin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Ben Wei
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Tushuai Li
- School of Biology and Food Engineering, Changshu Institute of Technology, Suzhou, 215500, People's Republic of China.
| |
Collapse
|
15
|
Wu X, Rong L, Tang R, Li Q, Wang F, Deng X, Miao L. Unveiling the role of CXCL10 in pancreatic cancer progression: A novel prognostic indicator. Open Med (Wars) 2025; 20:20241117. [PMID: 40129528 PMCID: PMC11931664 DOI: 10.1515/med-2024-1117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 11/14/2024] [Accepted: 11/25/2024] [Indexed: 03/26/2025] Open
Abstract
Objective Pancreatic cancer is distinguished by its high likelihood of metastasis and drug resistance, while the fundamental mechanisms are inadequately elucidated. This study aimed to identify pivotal hub genes associated with pancreatic cancer and assess their potential utility in predicting its onset and progression. Methods Weighted gene co-expression network analysis (WGCNA) combined with differential expression analysis identified novel susceptibility modules and hub genes for pancreatic cancer. Kyoto Encyclopedia of Genes and Genomes and gene ontology analyses were utilized to explore the potential roles of these hub genes. Receiver operator characteristic curves and nomogram models were developed to evaluate diagnostic efficacy. Mendelian randomization, flow cytometry, Transwell, and RNA sequencing were conducted to explore the association between C-X-C motif chemokine ligand 10 (CXCL10) and immune infiltration. Results WGCNA analysis was performed to build gene co-expression networks, and ten key genes were found. CXCL10 was the central gene, and its expression was significantly linked to the survival of patients with pancreatic cancer and their response to immune checkpoint inhibitors. CXCL10 demonstrated the ability to stimulate the differentiation of macrophages toward the M2 phenotype. CXCL10 could facilitate the metastasis of pancreatic cancer cells by modulating macrophage polarization. CXCL10 affects macrophage polarization by regulating the expression of vascular endothelial growth factor A. Conclusions CXCL10 demonstrates potential as a therapeutic target for managing pancreatic cancer.
Collapse
Affiliation(s)
- Xiaochao Wu
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Longfei Rong
- Department of General Surgery, SIR RUN RUN Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ruiyi Tang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Quanpeng Li
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fei Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xueting Deng
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lin Miao
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
16
|
Xiao Y, Sun S, Zheng N, Zhao J, Li X, Xu J, Li H, Du C, Zeng L, Zhang J, Yin X, Huang Y, Yang X, Yuan F, Jia X, Li B, Li B. Development of PDAC diagnosis and prognosis evaluation models based on machine learning. BMC Cancer 2025; 25:512. [PMID: 40114140 PMCID: PMC11924714 DOI: 10.1186/s12885-025-13929-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is difficult to detect early and highly aggressive, often leading to poor patient prognosis. Existing serum biomarkers like CA19-9 are limited in early diagnosis, failing to meet clinical needs. Machine learning (ML)/deep learning (DL) technologies have shown great potential in biomedicine. This study aims to establish PDAC differential diagnosis and prognosis assessment models using ML combined with serum biomarkers for early diagnosis, risk stratification, and personalized treatment recommendations, improving early diagnosis rates and patient survival. METHODS The study included serum biomarker data and prognosis information from 117 PDAC patients. ML models (Random Forest (RF), Neural Network (NNET), Support Vector Machine (SVM), and Gradient Boosting Machine (GBM)) were used for differential diagnosis, evaluated by accuracy, Kappa test, ROC curve, sensitivity, and specificity. COX proportional hazards model and DeepSurv DL model predicted survival risk, compared by C-index and Log-rank test. Based on DeepSurv's risk predictions, personalized treatment recommendations were made and their effectiveness assessed. RESULTS Effective PDAC diagnosis and prognosis models were built using ML. The validation set data shows that the accuracy of the RF, NNET, SVM, and GBM models are 84.21%, 84.21%, 76.97%, and 83.55%; the sensitivity are 91.26%, 90.29%, 89.32%, and 88.35%; and the specificity are 69.39%, 71.43%, 51.02%, and 73.47%. The Kappa values are 0.6266, 0.6307, 0.4336, and 0.6215; and the AUC are 0.889, 0.8488, 0.8488, and 0.8704, respectively. BCAT1, AMY, and CA12-5 were selected as modeling parameters for the prognosis model using COX regression. DeepSurv outperformed the COX model on both training and validation sets, with C-indexes of 0.738 and 0.724, respectively. The Kaplan-Meier survival curves indicate that personalized treatment recommendations based on DeepSurv can help patients achieve survival benefits. CONCLUSION This study built efficient PDAC diagnosis and prognosis models using ML, improving early diagnosis rates and prognosis accuracy. The DeepSurv model excelled in prognosis prediction and successfully guided personalized treatment recommendations and supporting PDAC clinical management.
Collapse
Affiliation(s)
- Yingqi Xiao
- Department of Clinical Laboratory, Beijing Electric Power Teaching Hospital, Capital Medical University, Beijing, China
| | - Shixin Sun
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Naxin Zheng
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Jing Zhao
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Xiaohan Li
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Jianmin Xu
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Haolian Li
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Chenran Du
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Lijun Zeng
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Juling Zhang
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Xiuyun Yin
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Yuan Huang
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Xuemei Yang
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Fang Yuan
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Xingwang Jia
- Department of Clinical Laboratory, Beijing Electric Power Teaching Hospital, Capital Medical University, Beijing, China.
| | - Boan Li
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China.
| | - Bo Li
- Department of Clinical Laboratory, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
17
|
Luo R, Li S, Yang C, Tang B, Li L, Luo C. Curcumin Inhibits the Development of Pancreatic Cancer by Targeting the circ_0079440/miR-522-3p/EIF4A1 Pathway. Cell Biochem Biophys 2025; 83:377-390. [PMID: 39102088 DOI: 10.1007/s12013-024-01466-8] [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] [Accepted: 07/25/2024] [Indexed: 08/06/2024]
Abstract
Pancreatic cancer (PC) is a common gastrointestinal cancer with high invasiveness and high mortality. Curcumin is a natural polyphenol with anti-tumor activity against different cancers, including PC. Curcumin has been verified to mediate the expression of circular RNAs (circRNAs) to inhibit tumor development. This study aimed to explore the function and regulatory mechanism of curcumin on circ_0079440 in PC. PC cells were treated with different concentrations of curcumin (0, 5, 10 or 15 μM) for 24 h. Gene expression in PC cells and tissues was detected using RT-qPCR. Cell malignant phenotypes were determined by functional assays. The levels of EMT-related proteins were tested using western blot. RNA interaction was determined using RNA pulldown assay, luciferase reporter assay and RIP assay. The results showed that curcumin suppressed cell proliferative, migratory, and invasive capabilities, and weakened epithelial-mesenchymal transition (EMT) in a concentration-dependent way. Circ_0079440 was expressed at a high level in PC and its level was reduced via curcumin administration in PC cells. Rescue assays showed that circ_0079440 overexpression reversed the suppressive effects of curcumin on PC cell malignant phenotypes. Furthermore, in the xenograft mouse models, curcumin treatment inhibited tumor growth and metastasis, and circ_0079440 upregulation reversed the function of curcumin. Additionally, circ_0079440 was revealed to bind to miR-522-3p to upregulate eukaryotic initiation factor 4A1 (EIF4A1) expression in PC cells. EIF4A1 expression was also downregulated by curcumin, and EIF4A1 overexpression abolished the suppressive functions of curcumin. Moreover, EIF4A overexpression or miR-522-3p inhibition counteracted the anti-tumor effects of circ_0079440 depletion on PC development. To sum up, curcumin suppresses PC development by targeting the circ_0079440/miR-522-3p/EIF4A1 pathway, which might provide novel therapeutic targets for treatment of PC.
Collapse
Affiliation(s)
- Ruiying Luo
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Shuang Li
- Department of respiratory medicine, The Third People's Hospital of Gansu Province, Lanzhou, 730000, Gansu, China
| | - Chi Yang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Baoyuan Tang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Long Li
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Changjiang Luo
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
18
|
Vazzano J, Chen W, Frankel WL. Intraoperative Frozen Section Evaluation of Pancreatic Specimens and Related Liver Lesions. Arch Pathol Lab Med 2025; 149:e63-e71. [PMID: 38736213 DOI: 10.5858/arpa.2023-0359-ra] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 05/14/2024]
Abstract
CONTEXT.— Frozen sections are essential in the surgical management of patients, especially those with pancreatic masses, because frozen sections can provide answers intraoperatively and aid in treatment decisions. Pancreas frozen sections are challenging because of the small tissue size, processing artifacts, neoadjuvant treatment effects, and concurrent pancreatitis-like obstructive changes. The authors present a review of intraoperative evaluation of pancreatic specimens. OBJECTIVE.— To provide an approach to the diagnosis of pancreatic adenocarcinoma on frozen sections and to discuss commonly encountered pitfalls. Indications for pancreas frozen sections and specific margin evaluation will be discussed. We will also review frozen section diagnosis of subcapsular liver lesions and tumors other than metastases of pancreatic ductal adenocarcinoma. DATA SOURCES.— Data sources included a literature review and the personal experiences of the authors. CONCLUSIONS.— The features for diagnosis of pancreatic adenocarcinoma include disordered architecture, glands at abnormal locations, and atypical cytology. It is important to be aware of the pitfalls and clues on frozen section. The evaluation of resection margins can be challenging, and in the setting of the resection of cystic tumors, the key is the diagnosis of high-grade dysplasia or cancer. Finally, it is vital to remember the differential diagnosis for subcapsular liver lesions because not all lesions will be metastases of adenocarcinomas or bile duct adenomas. Frozen sections remain a useful tool for the intraoperative management of patients with pancreatic tumors.
Collapse
Affiliation(s)
- Jennifer Vazzano
- From the Department of Pathology, The Ohio State University Wexner Medical Center, Columbus
| | - Wei Chen
- From the Department of Pathology, The Ohio State University Wexner Medical Center, Columbus
| | - Wendy L Frankel
- From the Department of Pathology, The Ohio State University Wexner Medical Center, Columbus
| |
Collapse
|
19
|
Guan X, Xu L, Liu J, Fei H, Wang C. Single-Cell Sequencing and Transcriptome Analysis Explored Changes in Midnolin-Related Immune Microenvironment and Constructed Combined Prognostic Model for Pancreatic Cancer. J Inflamm Res 2025; 18:2975-2990. [PMID: 40026303 PMCID: PMC11872096 DOI: 10.2147/jir.s503326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/22/2025] [Indexed: 03/05/2025] Open
Abstract
Background Pancreatic cancer has one of the worst prognoses of any malignant tumor. The value of MIDN, midnolin-related genes and midnolin-related immune infiltrating cells (MICs) in the prognosis of pancreatic cancer remains unknown. Methods Single-cell analysis were used to identify midnolin-related genes. Immune cell infiltration was obtained using CIBERSORT. The prognostic midnolin-related genes were identified through the utilization of Cox regression and the least absolute selection operator (LASSO) approach. The combined prognostic model was created using multifactorial Cox regression analysis. Survival analyses, immune microenvironment assessments, drug sensitivity checks were performed to evaluate the combined model performance. Finally, cellular experiments were carried out to confirm MIDN significance in pancreatic cancer. Results The combined model was constructed based on MIDN expression, prognostic model of 10 midnolin-related genes and M1 cell infiltration. Most immune checkpoint-related genes were expressed at greater levels in the low-risk group, suggesting a greater chance of immunotherapy's benefits. The most significant model gene, MIDN, was shown to have a function by cellular tests. In pancreatic cancer, MIDN knockdown drastically decreased pancreatic cancer cell lines' activity, proliferation, and invasive potential. Conclusion The combined model helped assess the prognosis of pancreatic cancer and offered fresh perspectives on immunotherapy in particular.
Collapse
Affiliation(s)
- Xiao Guan
- Department of Pancreatic and Gastric Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Lei Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Jinsong Liu
- Department of VIP Medical, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - He Fei
- Department of Pancreatic and Gastric Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Chengfeng Wang
- Department of Pancreatic and Gastric Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
20
|
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
|
21
|
Hu C, Chen Y, Yin X, Xu R, Yin C, Wang C, Zhao Y. Pancreatic endocrine and exocrine signaling and crosstalk in physiological and pathological status. Signal Transduct Target Ther 2025; 10:39. [PMID: 39948335 PMCID: PMC11825823 DOI: 10.1038/s41392-024-02098-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/20/2024] [Accepted: 12/03/2024] [Indexed: 02/16/2025] Open
Abstract
The pancreas, an organ with dual functions, regulates blood glucose levels through the endocrine system by secreting hormones such as insulin and glucagon. It also aids digestion through the exocrine system by secreting digestive enzymes. Complex interactions and signaling mechanisms between the endocrine and exocrine functions of the pancreas play a crucial role in maintaining metabolic homeostasis and overall health. Compelling evidence indicates direct and indirect crosstalk between the endocrine and exocrine parts, influencing the development of diseases affecting both. From a developmental perspective, the exocrine and endocrine parts share the same origin-the "tip-trunk" domain. In certain circumstances, pancreatic exocrine cells may transdifferentiate into endocrine-like cells, such as insulin-secreting cells. Additionally, several pancreatic diseases, including pancreatic cancer, pancreatitis, and diabetes, exhibit potential relevance to both endocrine and exocrine functions. Endocrine cells may communicate with exocrine cells directly through cytokines or indirectly by regulating the immune microenvironment. This crosstalk affects the onset and progression of these diseases. This review summarizes the history and milestones of findings related to the exocrine and endocrine pancreas, their embryonic development, phenotypic transformations, signaling roles in health and disease, the endocrine-exocrine crosstalk from the perspective of diseases, and potential therapeutic targets. Elucidating the regulatory mechanisms of pancreatic endocrine and exocrine signaling and provide novel insights for the understanding and treatment of diseases.
Collapse
Grants
- National High Level Hospital Clinical Research Funding (2022, 2022-PUMCH-D-001, to YZ), CAMS Innovation Fund for Medical Sciences (2021, 2021-I2M-1-002, to YZ), National Nature Science Foundation of China (2021, 82102810, to CW, the Fundamental Research Funds for the Central Universities(3332023123)
- cNational High Level Hospital Clinical Research Funding (2022, 2022-PUMCH-D-001, to YZ), CAMS Innovation Fund for Medical Sciences (2021, 2021-I2M-1-002, to YZ), National Nature Science Foundation of China (2021, 82102810, to CW, the Fundamental Research Funds for the Central Universities(3332023123)
Collapse
Affiliation(s)
- Chenglin Hu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Yuan Chen
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Xinpeng Yin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Chenxue Yin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Chengcheng Wang
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China.
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, PR China.
- Institute of Clinical Medicine, Peking Union Medical College Hospital, Beijing, PR China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China.
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, PR China.
| |
Collapse
|
22
|
Podină N, Gheorghe EC, Constantin A, Cazacu I, Croitoru V, Gheorghe C, Balaban DV, Jinga M, Țieranu CG, Săftoiu A. Artificial Intelligence in Pancreatic Imaging: A Systematic Review. United European Gastroenterol J 2025; 13:55-77. [PMID: 39865461 PMCID: PMC11866320 DOI: 10.1002/ueg2.12723] [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: 07/11/2024] [Revised: 10/24/2024] [Accepted: 11/03/2024] [Indexed: 01/28/2025] Open
Abstract
The rising incidence of pancreatic diseases, including acute and chronic pancreatitis and various pancreatic neoplasms, poses a significant global health challenge. Pancreatic ductal adenocarcinoma (PDAC) for example, has a high mortality rate due to late-stage diagnosis and its inaccessible location. Advances in imaging technologies, though improving diagnostic capabilities, still necessitate biopsy confirmation. Artificial intelligence, particularly machine learning and deep learning, has emerged as a revolutionary force in healthcare, enhancing diagnostic precision and personalizing treatment. This narrative review explores Artificial intelligence's role in pancreatic imaging, its technological advancements, clinical applications, and associated challenges. Following the PRISMA-DTA guidelines, a comprehensive search of databases including PubMed, Scopus, and Cochrane Library was conducted, focusing on Artificial intelligence, machine learning, deep learning, and radiomics in pancreatic imaging. Articles involving human subjects, written in English, and published up to March 31, 2024, were included. The review process involved title and abstract screening, followed by full-text review and refinement based on relevance and novelty. Recent Artificial intelligence advancements have shown promise in detecting and diagnosing pancreatic diseases. Deep learning techniques, particularly convolutional neural networks (CNNs), have been effective in detecting and segmenting pancreatic tissues as well as differentiating between benign and malignant lesions. Deep learning algorithms have also been used to predict survival time, recurrence risk, and therapy response in pancreatic cancer patients. Radiomics approaches, extracting quantitative features from imaging modalities such as CT, MRI, and endoscopic ultrasound, have enhanced the accuracy of these deep learning models. Despite the potential of Artificial intelligence in pancreatic imaging, challenges such as legal and ethical considerations, algorithm transparency, and data security remain. This review underscores the transformative potential of Artificial intelligence in enhancing the diagnosis and treatment of pancreatic diseases, ultimately aiming to improve patient outcomes and survival rates.
Collapse
Affiliation(s)
- Nicoleta Podină
- “Carol Davila” University of Medicine and PharmacyBucharestRomania
- Department of GastroenterologyPonderas Academic HospitalBucharestRomania
| | | | - Alina Constantin
- Department of GastroenterologyPonderas Academic HospitalBucharestRomania
| | - Irina Cazacu
- Oncology DepartmentFundeni Clinical InstituteBucharestRomania
| | - Vlad Croitoru
- Oncology DepartmentFundeni Clinical InstituteBucharestRomania
| | - Cristian Gheorghe
- “Carol Davila” University of Medicine and PharmacyBucharestRomania
- Center of Gastroenterology and HepatologyFundeni Clinical InstituteBucharestRomania
| | - Daniel Vasile Balaban
- “Carol Davila” University of Medicine and PharmacyBucharestRomania
- Department of Gastroenterology“Carol Davila” Central Military University Emergency HospitalBucharestRomania
| | - Mariana Jinga
- “Carol Davila” University of Medicine and PharmacyBucharestRomania
- Department of Gastroenterology“Carol Davila” Central Military University Emergency HospitalBucharestRomania
| | - Cristian George Țieranu
- “Carol Davila” University of Medicine and PharmacyBucharestRomania
- Department of Gastroenterology and HepatologyElias Emergency University HospitalBucharestRomania
| | - Adrian Săftoiu
- “Carol Davila” University of Medicine and PharmacyBucharestRomania
- Department of GastroenterologyPonderas Academic HospitalBucharestRomania
- Department of Gastroenterology and HepatologyElias Emergency University HospitalBucharestRomania
| |
Collapse
|
23
|
Jin D, Khan NU, Gu W, Lei H, Goel A, Chen T. Informatics strategies for early detection and risk mitigation in pancreatic cancer patients. Neoplasia 2025; 60:101129. [PMID: 39842383 PMCID: PMC11763847 DOI: 10.1016/j.neo.2025.101129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/17/2025] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
This review provides a comprehensive overview of the current landscape in pancreatic cancer (PC) screening, diagnosis, and early detection. This emphasizes the need for targeted screening in high-risk groups, particularly those with familial predispositions and genetic mutations, such as BRCA1, BRCA2, and PALB2. This review highlights the sporadic nature of most PC cases and significant risk factors, including smoking, alcohol consumption, obesity, and diabetes. Advanced imaging techniques, such as Endoscopic Ultrasound (EUS) and Contrast-Enhanced Harmonic Imaging (CEH-EUS), have been discussed for their superior sensitivity in early detection. This review also explores the potential of novel biomarkers, including those found in body fluids, such as serum, plasma, urine, and bile, as well as the emerging role of liquid biopsy technologies in analyzing circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and exosomes. AI-driven approaches, such as those employed in Project Felix and CancerSEEK, have been highlighted for their potential to enhance early detection through deep learning and biomarker discovery. This review underscores the importance of universal genetic testing and the integration of AI with traditional diagnostic methods to improve outcomes in high-risk individuals. Additionally, this review points to future directions in PC diagnostics, including next-generation imaging, molecular biomarkers, and personalized medicine, aiming to overcome current diagnostic challenges and improve survival rates. Ultimately, the review advocates the adoption of informatics and AI-driven strategies to enhance early detection, reduce morbidity, and save lives in the fight against pancreatic cancer.
Collapse
Affiliation(s)
- Di Jin
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, China; Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Najeeb Ullah Khan
- Institute of Biotechnology & Genetic Engineering (Health Division), The University of Agriculture Peshawar, Peshawar, PO Box 25130, Pakistan.
| | - Wei Gu
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, China; Wenzhou Medical University, Wenzhou, 325000, China.
| | - Huijun Lei
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, China.
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California, USA; City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| | - Tianhui Chen
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, China.
| |
Collapse
|
24
|
Balaraman AK, Moglad E, Afzal M, Babu MA, Goyal K, Roopashree R, Kaur I, Kumar S, Kumar MR, Chauhan AS, Hemalatha S, Gupta G, Ali H. Liquid biopsies and exosomal ncRNA: Transforming pancreatic cancer diagnostics and therapeutics. Clin Chim Acta 2025; 567:120105. [PMID: 39706249 DOI: 10.1016/j.cca.2024.120105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Pancreatic cancer is a highly fatal malignancy due to poor early detection rate and resistance to conventional therapies. This review examines the potential for liquid biopsy as a transformative technology to identify diagnostic and therapeutic targets in pancreatic cancer. Specifically, we explore emerging biomarkers such as exosomal non-coding RNAs (ncRNAs), circulating tumor DNA (ctDNA), and circulating tumor cells (CTCs). Tumor-derived exosomes contain nucleic acid and protein that reflect the unique molecular landscape of the malignancy and can serve as an alternative diagnostic approach vs traditional biomarkers like CA19-9. Herein we highlight exosomal miRNAs, lncRNAs, and other ncRNAs alongside ctDNA and CTC-based strategies, evaluating their combined ability to improve early detection, disease monitoring and treatment response. Furthermore, the therapeutic implications of ncRNAs such as lncRNA UCA1 and miR-3960 in chemoresistance and progression are also discussed via suppression of EZH2 and PTEN/AKT pathways. Emerging therapeutic strategies that target the immune response, epithelial-mesenchymal transition (EMT) and drug resistance are explored. This review demonstrates a paradigm shift in pancreatic cancer management toward personalized, less invasive and more effective approaches.
Collapse
Affiliation(s)
- Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - MRavi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research and Innovation, Uttaranchal University, India
| | - S Hemalatha
- Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Porur, Chennai, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| |
Collapse
|
25
|
e Silva DRM, de Oliveira MM, Fernandes GA, Curado MP. The burden of pancreatic cancer in Latin America and the Caribbean: trends in incidence, mortality and DALYs from 1990 to 2019. Ecancermedicalscience 2025; 19:1827. [PMID: 40177150 PMCID: PMC11959134 DOI: 10.3332/ecancer.2025.1827] [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: 03/23/2024] [Indexed: 04/05/2025] Open
Abstract
This study analyzed the burden of pancreatic cancer by investigating its incidence, mortality and disability-adjusted life years (DALYs), as well as the proportion of pancreatic cancer deaths attributable to behavioural and metabolic risk factors in Latin America and the Caribbean (LAC) countries. Methods This study focuses on pancreatic cancer using the Global Burden of Disease 2019 study database. Results were described for 23 LAC countries for 1990-2019, evaluating their age-standardised incidence rates, mortality rates, DALYs, average annual percent change and the fraction of deaths attributable to behavioural and metabolic risk factors. Results We observed that in LAC, pancreatic cancer incidence rates ranged from 1.2 in Haiti to 15.8/100,000 in Uruguay among men. The highest increase in incidence rate was observed in Trinidad and Tobago: 7.7% per year. The mortality rate was higher in Uruguay and lower in Haiti, for both sexes. The highest rise in the numbers of DALYs in 2019 was observed in Brazil and Mexico. The proportion of pancreatic cancer deaths attributable to smoking was reduced between 1990 and 2019 for both sexes in LAC countries; however, it increased for metabolic risk factors. Conclusion The increasing trend in pancreatic cancer observed in LAC may be associated with a rise in risk factors such as high fasting plasma glucose and high body mass index in both sexes. This trend will likely have a substantial impact on the healthcare system in the coming decades.
Collapse
Affiliation(s)
- Diego Rodrigues Mendonça e Silva
- Postgraduate Program in Epidemiology, School of Public Health, University of São Paulo, São Paulo, SP 01246-904, Brazil
- Hospital Cancer Registry, A.C.Camargo Cancer Center, São Paulo, SP 01246-904, Brazil
- https://orcid.org/0000-0001-8469-8415
| | - Max Moura de Oliveira
- Department of Collective Health, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiania, GO 01246-904, Brazil
- https://orcid.org/0000-0002-0804-5145
| | - Gisele Aparecida Fernandes
- Group of Epidemiology and Statistics on Cancer, A.C.Camargo Cancer Center, São Paulo, SP 01246-904, Brazil
- https://orcid.org/0000-0002-5978-3279
| | - Maria Paula Curado
- Postgraduate Program in Epidemiology, School of Public Health, University of São Paulo, São Paulo, SP 01246-904, Brazil
- Hospital Cancer Registry, A.C.Camargo Cancer Center, São Paulo, SP 01246-904, Brazil
- Group of Epidemiology and Statistics on Cancer, A.C.Camargo Cancer Center, São Paulo, SP 01246-904, Brazil
- https://orcid.org/0000-0001-8172-2483
| |
Collapse
|
26
|
Luo YG, Wu M, Chen HG. Retrospective analysis of pathological types and imaging features in pancreatic cancer: A comprehensive study. World J Gastrointest Oncol 2025; 17:99153. [PMID: 39817138 PMCID: PMC11664627 DOI: 10.4251/wjgo.v17.i1.99153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Pancreatic cancer remains one of the most lethal malignancies worldwide, with a poor prognosis often attributed to late diagnosis. Understanding the correlation between pathological type and imaging features is crucial for early detection and appropriate treatment planning. AIM To retrospectively analyze the relationship between different pathological types of pancreatic cancer and their corresponding imaging features. METHODS We retrospectively analyzed the data of 500 patients diagnosed with pancreatic cancer between January 2010 and December 2020 at our institution. Pathological types were determined by histopathological examination of the surgical specimens or biopsy samples. The imaging features were assessed using computed tomography, magnetic resonance imaging, and endoscopic ultrasound. Statistical analyses were performed to identify significant associations between pathological types and specific imaging characteristics. RESULTS There were 320 (64%) cases of pancreatic ductal adenocarcinoma, 75 (15%) of intraductal papillary mucinous neoplasms, 50 (10%) of neuroendocrine tumors, and 55 (11%) of other rare types. Distinct imaging features were identified in each pathological type. Pancreatic ductal adenocarcinoma typically presents as a hypodense mass with poorly defined borders on computed tomography, whereas intraductal papillary mucinous neoplasms present as characteristic cystic lesions with mural nodules. Neuroendocrine tumors often appear as hypervascular lesions in contrast-enhanced imaging. Statistical analysis revealed significant correlations between specific imaging features and pathological types (P < 0.001). CONCLUSION This study demonstrated a strong association between the pathological types of pancreatic cancer and imaging features. These findings can enhance the accuracy of noninvasive diagnosis and guide personalized treatment approaches.
Collapse
Affiliation(s)
- Yang-Gang Luo
- Pathology Department, Xuanhan County People’s Hospital, Dazhou 636150, Sichuan Province, China
| | - Mei Wu
- Pathology Department, Xuanhan County People’s Hospital, Dazhou 636150, Sichuan Province, China
| | - Hong-Guang Chen
- Pathology Department, Xuanhan County People’s Hospital, Dazhou 636150, Sichuan Province, China
| |
Collapse
|
27
|
Puik JR, Poels TT, Hooijer GKJ, Cysouw MCF, Verheij J, Wilmink JW, Giovannetti E, Kazemier G, Sarasqueta AF, Oprea-Lager DE, Swijnenburg RJ. 18F-Prostate-Specific Membrane Antigen PET/CT imaging for potentially resectable pancreatic cancer (PANSCAN-2): a phase I/II study. Cancer Imaging 2025; 25:2. [PMID: 39810252 PMCID: PMC11734402 DOI: 10.1186/s40644-025-00822-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Current diagnostic imaging modalities have limited ability to differentiate between malignant and benign pancreaticobiliary disease, and lack accuracy in detecting lymph node metastases. 18F-Prostate-Specific Membrane Antigen (PSMA) PET/CT is an imaging modality used for staging of prostate cancer, but has incidentally also identified PSMA-avid pancreatic lesions, histologically characterized as pancreatic ductal adenocarcinoma (PDAC). This phase I/II study aimed to assess the feasibility of 18F-PSMA PET/CT to detect PDAC. METHODS Seventeen patients with clinically resectable PDAC underwent 18F-PSMA PET/CT prior to surgical resection. Images were analyzed both visually and (semi)quantitatively by deriving the maximum standardized uptake value (SUVmax) and tumor-to-background ratio (TBR). TBR was defined as the ratio between SUVmax of the primary tumor divided by SUVmax of the aortic blood pool. Finally, tracer uptake on PET was correlated to tissue expression of PSMA in surgical specimens. RESULTS Out of 17 PSMA PET/CT scans, 13 scans demonstrated positive PSMA tracer uptake, with a mean SUVmax of 5.0 ± 1.3. The suspected primary tumor was detectable (TBR ≥ 2) with a mean TBR of 3.3 ± 1.3. For histologically confirmed PDAC, mean SUVmax and mean TBR were 4.9 ± 1.2 and 3.3 ± 1.5, respectively. Although eight patients had histologically confirmed regional lymph node metastases and two patients had distant metastases, none of these metastases demonstrated 18F-PSMA uptake. There was no correlation between 18F-PSMA PET/CT SUVmax and tissue expression of PSMA in surgical specimens. CONCLUSIONS 18F-PSMA PET/CT was able to detect several pancreaticobiliary cancers, including PDAC. However, uptake was generally low, not specific to PDAC and no tracer uptake was observed in lymph node or distant metastases. The added value of PSMA PET in this setting appears to be limited. TRIAL REGISTRATION The trial is registered as PANSCAN-2 in the European Clinical Trials Database (EudraCT number: 2020-002185-14).
Collapse
Affiliation(s)
- Jisce R Puik
- Department of Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Thomas T Poels
- Department of Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Gerrit K J Hooijer
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Pathology, UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Matthijs C F Cysouw
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Joanne Verheij
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Pathology, UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Johanna W Wilmink
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Elisa Giovannetti
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, Pisa, Italy
| | - Geert Kazemier
- Department of Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Arantza Farina Sarasqueta
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Pathology, UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Daniela E Oprea-Lager
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Rutger-Jan Swijnenburg
- Department of Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands.
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
- Department of Surgery, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands.
| |
Collapse
|
28
|
Jogi M, Asnani H, Singh S, Kumar P. Nimbolide: A Potential Phytochemical Agent in Multimodal Pancreatic Cancer Therapies. Mini Rev Med Chem 2025; 25:27-41. [PMID: 38874049 DOI: 10.2174/0113895575293138240527061556] [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: 12/19/2023] [Revised: 04/09/2024] [Accepted: 04/20/2024] [Indexed: 06/15/2024]
Abstract
A significant contributor to cancer-related death, pancreatic cancer (PC) has a terrible prognosis in general that has not altered over many years. Currently, it is extremely difficult to prevent disease or discover it early enough to initiate treatment. PC is a challenging malignancy to treat, and several major impediments significantly impact the effectiveness of its treatment. These obstacles primarily include chemoresistance, drug toxicity, and limited drug bioavailability. Phytochemicals can be used as an alternative to chemotherapeutic drugs, or they can augment the anticancer properties of the chemotherapeutic agents. Nimbolide (NL) is a prominent limonoid compound found in Azadirachta indica, and has garnered substantial attention as a phytochemical with anticancer potential. It has powerful antiproliferative effects on a variety of cancer cell lines and is effective as a chemotherapeutic in preclinical studies. The primary modes of action of NL include suppression of metastasis and angiogenesis, activation of apoptosis, anti-proliferation, and control of enzymes that metabolize carcinogens. Despite numerous pharmacodynamic (PD) investigations, NL is still in the early stages of the drug development process because no comprehensive pharmacokinetic studies or long-term toxicity studies. Preclinical and toxicological assessments should be conducted to establish an appropriate dosage range, ensuring the safety of NL for its application in initial human clinical trials. This review endeavors to provide a comprehensive summary of the current developmental stage of NL along with nanoparticles as a principal candidate for therapeutic purposes in PC.
Collapse
Affiliation(s)
- Mukesh Jogi
- Division of Molecular Biology, ICMR-National Institute of Cancer Prevention and Research (NICPR), NOIDA, India
- Amity Institute of Biotechnology, Amity University, NOIDA, India
- Department of Biotecnology ICMR- National Institute for Research in Environmental Health, Bhopal, India
| | - Hitakshi Asnani
- Banasthli Vidyapith, Radha Kishnpura, Rajasthan, 304022, India
| | - Sohini Singh
- Amity Institute of Biotechnology, Amity University, NOIDA, India
| | - Pramod Kumar
- Division of Molecular Biology, ICMR-National Institute of Cancer Prevention and Research (NICPR), NOIDA, India
| |
Collapse
|
29
|
Lee JH, Lee CG, Kim MS, Kim S, Song M, Zhang H, Yang E, Kwon YH, Jung YH, Hyeon DY, Choi YJ, Oh S, Joe DJ, Kim TS, Jeon S, Huang Y, Kwon TH, Lee KJ. Deeply Implantable, Shape-Morphing, 3D MicroLEDs for Pancreatic Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2411494. [PMID: 39679727 DOI: 10.1002/adma.202411494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/07/2024] [Indexed: 12/17/2024]
Abstract
Controlled photooxidation-mediated disruption of collagens in the tumor microenvironment can reduce desmoplasia and enhance immune responsiveness. However, achieving effective light delivery to solid tumors, particularly those with dynamic volumetric changes like pancreatic ductal adenocarcinoma (PDAC), remains challenging and limits the repeated and sustained photoactivation of drugs. Here, 3D, shape-morphing, implantable photonic devices (IPDs) are introduced that enable tumor-specific and continuous light irradiation for effective metronomic photodynamic therapy (mPDT). This IPD adheres seamlessly to the surface of orthotopic PDAC tumors, mitigating issues related to mechanical mismatch, delamination, and internal lesions. In freely moving mouse models, mPDT using the IPD with close adhesion significantly reduces desmoplastic tumor volume without causing cytotoxic effects in healthy tissues. These promising in vivo results underscore the potential of an adaptable and unidirectional IPD design in precisely targeting cancerous organs, suggesting a meaningful advance in light-based therapeutic technologies.
Collapse
Affiliation(s)
- Jae Hee Lee
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA
| | - Chae Gyu Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Min Seo Kim
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Seungyeob Kim
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Myoung Song
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Haohui Zhang
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Eunbyeol Yang
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Yoon Hee Kwon
- O2MEDi Incorporation, Ulsan, 44919, Republic of Korea
| | - Young Hoon Jung
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Dong Yeol Hyeon
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Yoon Ji Choi
- In Vivo Research Center, UNIST Central Research Facilities, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Seyong Oh
- Division of Electrical Engineering, Hanyang University ERICA, Ansan, 15588, Republic of Korea
| | - Daniel J Joe
- Division of Biomedical Metrology, Korea Research Institute of Standards and Science (KRISS), Daejeon, 34113, Republic of Korea
| | - Taek-Soo Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Sanghun Jeon
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Yonggang Huang
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA
- Departments of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Tae-Hyuk Kwon
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- O2MEDi Incorporation, Ulsan, 44919, Republic of Korea
| | - Keon Jae Lee
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| |
Collapse
|
30
|
Danpanichkul P, Suparan K, Tothanarungroj P, Dejvajara D, Rakwong K, Pang Y, Barba R, Thongpiya J, Fallon MB, Harnois D, Lui RN, Wallace MB, Yang JD, Roberts LR, Wijarnpreecha K. Epidemiology of gastrointestinal cancers: a systematic analysis from the Global Burden of Disease Study 2021. Gut 2024; 74:26-34. [PMID: 39242191 DOI: 10.1136/gutjnl-2024-333227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/21/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Gastrointestinal cancers comprise nearly one-third of global mortality from cancer, yet the comprehensive global burden of these cancers remains uninvestigated. OBJECTIVE We aimed to assess the global, regional and national burden of gastrointestinal cancers. DESIGNS Data on oesophagus, gastric, colorectal, liver, pancreas and biliary tract cancers were extracted from the Global Burden of Disease 2021 database. Age-standardised incidence rate (ASIR) and age-standardised death rate (ASDR) were calculated by sex, region and Sociodemographic Index (SDI). RESULTS In 2021, there were 5.26 million incidences and 3.70 million deaths from gastrointestinal cancer. The greatest burden is from colorectal, followed by gastric, oesophageal, pancreatic, liver and biliary tract cancer. We noted geographical and socioeconomic differences in ASIR and ASDR across all types of cancers. From 2000 to 2021, ASIR increased for colorectal cancer (annual percent change (APC): 0.10%, 95% CI 0.05% to 0.14%), pancreatic cancer (APC: 0.27%, 95% CI 0.14% to 0.41%), and liver cancer from metabolic dysfunction-associated steatotic liver disease (APC: 0.62%, 95% CI 0.58% to 0.67%) and alcohol-related liver disease (APC: 0.26%, 95% CI 0.22% to 0.30%). ASDR increased for pancreatic cancer (APC: 0.18%, 95% CI 0.02% to 0.34%). Higher SDI countries had higher incidence rates for most types of gastrointestinal cancer. CONCLUSIONS Although the ASIR of oesophageal, gastric and biliary tract cancer has decreased, the ASIR still increased in colorectal, pancreatic and liver cancer from steatotic liver disease. Public policies are important for controlling gastrointestinal cancers-most importantly, reducing alcohol consumption, hepatitis B immunisation and tackling the burden of metabolic diseases.
Collapse
Affiliation(s)
- Pojsakorn Danpanichkul
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | | | | | | | | | - Yanfang Pang
- Department of Microbiology, Chiang Mai University, Chiang Mai, Thailand
- Affiliated Hospital of Youjiang Medical University of Nationalities, Baise, Guangxi, People's Republic of China
- National Immunological Laboratory for Traditional Chinese Medicine, Baise, Guangxi, People's Republic of China
- Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi, Guangxi, Guangxi, People's Republic of China
| | - Romelia Barba
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Jerapas Thongpiya
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Michael B Fallon
- Division of Gastroenterology and Hepatology, Department of Medicine, The University of Arizona College of Medicine Phoenix, Phoenix, Arizona, USA
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Banner - University Medical Center Phoenix, Phoenix, Arizona, USA
| | - Denise Harnois
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida, USA
| | - Rashid N Lui
- Institute of Digestive Disease, Chinese University of Hong Kong, New Territories, Hong Kong, People's Republic of China
| | - Michael B Wallace
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida, USA
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Comprehensive Transplant Center, and Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Lewis R Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Karn Wijarnpreecha
- Division of Gastroenterology and Hepatology, Department of Medicine, The University of Arizona College of Medicine Phoenix, Phoenix, Arizona, USA
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Banner - University Medical Center Phoenix, Phoenix, Arizona, USA
- Mayo Clinic Florida, Jacksonville, Florida, USA
| |
Collapse
|
31
|
Pietsch FL, Haag F, Ayx I, Grawe F, Vellala AK, Schoenberg SO, Froelich MF, Tharmaseelan H. Textural heterogeneity of liver lesions in CT imaging - comparison of colorectal and pancreatic metastases. Abdom Radiol (NY) 2024; 49:4295-4306. [PMID: 39115682 PMCID: PMC11522118 DOI: 10.1007/s00261-024-04511-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE Tumoral heterogeneity poses a challenge for personalized cancer treatments. Especially in metastasized cancer, it remains a major limitation for successful targeted therapy, often leading to drug resistance due to tumoral escape mechanisms. This work explores a non-invasive radiomics-based approach to capture textural heterogeneity in liver lesions and compare it between colorectal cancer (CRC) and pancreatic cancer (PDAC). MATERIALS AND METHODS In this retrospective single-center study 73 subjects (42 CRC, 31 PDAC) with 1291 liver metastases (430 CRC, 861 PDAC) were segmented fully automated on contrast-enhanced CT images by a UNet for medical images. Radiomics features were extracted using the Python package Pyradiomics. The mean coefficient of variation (CV) was calculated patient-wise for each feature to quantify the heterogeneity. An unpaired t-test identified features with significant differences in feature variability between CRC and PDAC metastases. RESULTS In both colorectal and pancreatic liver metastases, interlesional heterogeneity in imaging can be observed using quantitative imaging features. 75 second-order features were extracted to compare the varying textural characteristics. In total, 18 radiomics features showed a significant difference (p < 0.05) in their expression between the two malignancies. Out of these, 16 features showed higher levels of variability within the cohort of pancreatic metastases, which, as illustrated in a radar plot, suggests greater textural heterogeneity for this entity. CONCLUSIONS Radiomics has the potential to identify the interlesional heterogeneity of CT texture among individual liver metastases. In this proof-of-concept study for the quantification and comparison of imaging-related heterogeneity in liver metastases a variation in the extent of heterogeneity levels in CRC and PDAC liver metastases was shown.
Collapse
Affiliation(s)
- Friedrich L Pietsch
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Florian Haag
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Isabelle Ayx
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Freba Grawe
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Abhinay K Vellala
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Stefan O Schoenberg
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Matthias F Froelich
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Hishan Tharmaseelan
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|
32
|
Saha B, Chakravarty S, Ray S, Saha H, Das K, Ghosh I, Mallick B, Biswas NK, Goswami S. Correlating tissue and plasma‑specific piRNA changes to predict their possible role in pancreatic malignancy and chronic inflammation. Biomed Rep 2024; 21:186. [PMID: 39420923 PMCID: PMC11484194 DOI: 10.3892/br.2024.1874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
The aggressiveness of pancreatic ductal adenocarcinoma is primarily due to lack of effective early detection biomarkers. Circulating non-coding RNAs serve as diagnostic or prognostic biomarkers in multiple types of cancer. Comparison of their expression between diseased tissue and relevant body fluids such as saliva, urine, bile, pancreatic juice, blood etc. may reveal mechanistic involvement of common non-coding RNAs. piwi-interacting RNAs (piRNAs) are a class of non-coding RNAs. The aim of the present study was to investigate plasma and tumour tissue piRNA changes in patients with pancreatic cancer (PC) and explore the possible role in tumorigenesis and pancreatic inflammation. Sequencing of circulating plasma small RNAs from patients with PC and chronic pancreatitis (CP) was performed and differentially expressed piRNAs were compared with those in tissues. Subsequent search for target genes for those piRNAs was performed followed by pathway and cluster analysis. A total of 36 piRNAs were shown to be deregulated in pancreatic tumour tissue and alteration of 11 piRNAs was detected in plasma of patients with PC. piRNAs hsa-piR-23246, hsa-piR-32858 and hsa-piR-9137 may serve a key role in PC development as their expression was correlated in both plasma and tumour tissue. Key piRNA-target interactions interfering with key biological pathways were also characterized. A total of 19 deregulated piRNAs in plasma samples of patients with CP was identified; these targeted genes responsible for chronic inflammation. Therefore, the present study provides a comprehensive description of piRNA alteration in pancreatic malignancy and inflammation; these may be explored for biomarker potential in future.
Collapse
Affiliation(s)
- Barsha Saha
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, Nadia, West Bengal 741251, India
- Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad 121001, India
| | - Shouvik Chakravarty
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, Nadia, West Bengal 741251, India
- Regional Centre for Biotechnology, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad 121001, India
| | - Sukanta Ray
- Institute of Postgraduate Medical Education & Research, Kolkata 700020, India
| | - Hemabha Saha
- Institute of Postgraduate Medical Education & Research, Kolkata 700020, India
| | - Kshaunish Das
- Institute of Postgraduate Medical Education & Research, Kolkata 700020, India
| | - Indranil Ghosh
- Chittaranjan National Cancer Institute, Kolkata 700026, India
| | | | - Nidhan K. Biswas
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, Nadia, West Bengal 741251, India
| | - Srikanta Goswami
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, Nadia, West Bengal 741251, India
| |
Collapse
|
33
|
Yang W, Hu P, Zuo C. Application of imaging technology for the diagnosis of malignancy in the pancreaticobiliary duodenal junction (Review). Oncol Lett 2024; 28:596. [PMID: 39430731 PMCID: PMC11487531 DOI: 10.3892/ol.2024.14729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
The pancreaticobiliary duodenal junction (PBDJ) is the connecting area of the pancreatic duct, bile duct and duodenum. In a broad sense, it refers to a region formed by the head of the pancreas, the pancreatic segment of the common bile duct and the intraduodenal segment, the descending and the horizontal part of the duodenum, and the soft tissue around the pancreatic head. In a narrow sense, it refers to the anatomical Vater ampulla. Due to its complex and variable anatomical features, and the diversity of pathological changes, it is challenging to make an early diagnosis of malignancy at the PBDJ and define the histological type. The unique anatomical structure of this area may be the basis for the occurrence of malignant tumors. Therefore, understanding and subclassifying the anatomical configuration of the PBDJ is of great significance for the prevention and treatment of malignant tumors at their source. The present review comprehensively discusses commonly used imaging techniques and other new technologies for diagnosing malignancy at the PBDJ, offering evidence for physicians and patients to select appropriate examination methods.
Collapse
Affiliation(s)
- Wanyi Yang
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
- Graduates Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410013, P.R. China
| | - Pingsheng Hu
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
| | - Chaohui Zuo
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
- Graduates Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
34
|
Javed S, Qureshi TA, Wang L, Azab L, Gaddam S, Pandol SJ, Li D. An insight to PDAC tumor heterogeneity across pancreatic subregions using computed tomography images. Front Oncol 2024; 14:1378691. [PMID: 39600638 PMCID: PMC11588633 DOI: 10.3389/fonc.2024.1378691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is an exceptionally deadly form of pancreatic cancer with an extremely low survival rate. From diagnosis to treatment, PDAC is highly challenging to manage. Studies have demonstrated that PDAC tumors in distinct regions of the pancreas exhibit unique characteristics, influencing symptoms, treatment responses, and survival rates. Gaining insight into the heterogeneity of PDAC tumors based on their location in the pancreas can significantly enhance overall management of PDAC. Previous studies have explored PDAC tumor heterogeneity across pancreatic subregions based on their genetic and molecular profiles through biopsy-based histologic assessment. However, biopsy examinations are highly invasive and impractical for large populations. Abdominal imaging, such as Computed Tomography (CT) offers a completely non-invasive means to evaluate PDAC tumor heterogeneity across pancreatic subregions and an opportunity to correlate image feature of tumors with treatment outcome and monitoring. In this study, we explored the inter-tumor heterogeneity in PDAC tumors across three primary pancreatic subregions: the head, body, and tail. Utilizing contrast-enhanced abdominal CT scans and a thorough radiomic analysis of PDAC tumors, several morphological and textural tumor features were identified to be notably different between tumors in the head and those in the body and tail regions. To validate the significance of the identified features, a machine learning ML model was trained to automatically classify PDAC tumors into their respective regions i.e. head or body/tail subregion using their CT features. The study involved 200 CT abdominal scans, with 100 used for radiomic analysis and model training, and the remaining 100 for model testing. The ML model achieved an average classification accuracy, sensitivity, and specificity of 87%, 86%, and 88% on the testing scans respectively. Evaluating the heterogeneity of PDAC tumors across pancreatic subregions provides valuable insights into tumor composition and has the potential to enhance diagnosis and personalize treatment based on tumor characteristics and location.
Collapse
Affiliation(s)
- Sehrish Javed
- Cedars Sinai Medical Center, Los Angeles, CA, United States
| | | | | | | | | | | | - Debiao Li
- Cedars Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
35
|
Kotb A, Hafeji Z, Jesry F, Lintern N, Pathak S, Smith AM, Lutchman KRD, de Bruin DM, Hurks R, Heger M, Khaled YS. Intra-Operative Tumour Detection and Staging in Pancreatic Cancer Surgery: An Integrative Review of Current Standards and Future Directions. Cancers (Basel) 2024; 16:3803. [PMID: 39594758 PMCID: PMC11592681 DOI: 10.3390/cancers16223803] [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: 06/09/2024] [Revised: 10/15/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Surgical resection for pancreatic ductal adenocarcinoma (PDAC) entails the excision of the primary tumour and regional lymphadenectomy. This traditional strategy is challenged by the high rate of early recurrence, suggesting inadequate disease staging. Novel methods of intra-operative staging are needed to allow surgical resection to be tailored to the disease's biology. METHODS A search of published articles on the PubMed and Embase databases was performed using the terms 'pancreas' OR 'pancreatic' AND 'intra-operative staging/detection' OR 'guided surgery'. Articles published between January 2000 and June 2023 were included. Technologies that offered intra-operative staging and tailored treatment were curated and summarised in the following integrative review. RESULTS lymph node (LN) mapping and radioimmunoguided surgery have shown promising results but lacked practicality to facilitate real-time intra-operative staging for PDAC. Fluorescence-guided surgery (FGS) offers high contrast and sensitivity, enabling the identification of cancerous tissue and positive LNs with improved precision following intravenous administration of a fluorescent agent. The unique properties of optical coherence tomography and ultrasound elastography lend themselves to be platforms for virtual biopsy intra-operatively. CONCLUSIONS Accurate intra-operative staging of PDAC, localisation of metastatic LNs, and identification of extra-pancreatic disease remain clinically unmet needs under current detection methods and staging standards. Tumour-specific FGS combined with other diagnostic and therapeutic modalities could improve tumour detection and staging in patients with PDAC.
Collapse
Affiliation(s)
- Ahmed Kotb
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| | - Zaynab Hafeji
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| | - Fadel Jesry
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| | - Nicole Lintern
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| | - Samir Pathak
- The Pancreato-Biliary Unit, St James’s University Teaching Hospital, Leeds LS9 7TF, UK
| | - Andrew M. Smith
- The Pancreato-Biliary Unit, St James’s University Teaching Hospital, Leeds LS9 7TF, UK
| | - Kishan R. D. Lutchman
- Department of Surgery, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands
- Department of Biomedical Engineering and Physics, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands
| | - Daniel M. de Bruin
- Department of Biomedical Engineering and Physics, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands
| | - Rob Hurks
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China
| | - Yazan S. Khaled
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
- The Pancreato-Biliary Unit, St James’s University Teaching Hospital, Leeds LS9 7TF, UK
| |
Collapse
|
36
|
Qadir RMAB, Umair MB, Tariq UB, Ahmad A, Kiran W, Shahid MH. Unraveling Pancreatic Cancer: Epidemiology, Risk Factors, and Global Trends. Cureus 2024; 16:e72816. [PMID: 39493341 PMCID: PMC11528318 DOI: 10.7759/cureus.72816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2024] [Indexed: 11/05/2024] Open
Abstract
Pancreatic cancer is one of the most lethal malignancies, characterized by late diagnosis, rapid progression, and limited treatment options. This literature review comprehensively examines the epidemiology, risk factors, diagnostic challenges, treatment modalities, and prognosis of pancreatic cancer. It highlights the global disparities in incidence and outcomes, exploring the influence of socioeconomic, environmental, and genetic factors on disease progression. In addition, this review discusses recent advancements in diagnostic tools and treatment strategies, identifying gaps in current research and clinical practices. The synthesis aims to inform future research directions and policy-making efforts to reduce the global burden of pancreatic cancer and improve patient outcomes.
Collapse
Affiliation(s)
| | | | - Umar Bin Tariq
- General Surgery, Southmead Hospital Bristol, North Bristol NHS Trust, Bristol, GBR
| | - Arslan Ahmad
- Emergency Medicine, Weston General Hospital, University Hospitals Bristol and Weston NHS Foundation Trust, Weston-super-Mare, GBR
| | - Wajeeha Kiran
- Trauma and Orthopaedics, Morriston Hospital, Swansea, GBR
| | | |
Collapse
|
37
|
Siciliano AC, Forciniti S, Onesto V, Iuele H, Cave DD, Carnevali F, Gigli G, Lonardo E, Del Mercato LL. A 3D Pancreatic Cancer Model with Integrated Optical Sensors for Noninvasive Metabolism Monitoring and Drug Screening. Adv Healthc Mater 2024; 13:e2401138. [PMID: 38978424 DOI: 10.1002/adhm.202401138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/06/2024] [Indexed: 07/10/2024]
Abstract
A distinct feature of pancreatic ductal adenocarcinoma (PDAC) is a prominent tumor microenvironment (TME) with remarkable cellular and spatial heterogeneity that meaningfully impacts disease biology and treatment resistance. The dynamic crosstalk between cancer cells and the dense stromal compartment leads to spatially and temporally heterogeneous metabolic alterations, such as acidic pH that contributes to drug resistance in PDAC. Thus, monitoring the extracellular pH metabolic fluctuations within the TME is crucial to predict and to quantify anticancer drug efficacy. Here, a simple and reliable alginate-based 3D PDAC model embedding ratiometric optical pH sensors and cocultures of tumor (AsPC-1) and stromal cells for simultaneously monitoring metabolic pH variations and quantify drug response is presented. By means of time-lapse confocal laser scanning microscopy (CLSM) coupled with a fully automated computational analysis, the extracellular pH metabolic variations are monitored and quantified over time during drug testing with gemcitabine, folfirinox, and paclitaxel, commonly used in PDAC therapy. In particular, the extracellular acidification is more pronounced after drugs treatment, resulting in increased antitumor effect correlated with apoptotic cell death. These findings highlight the importance of studying the influence of cellular metabolic mechanisms on tumor response to therapy in 3D tumor models, this being crucial for the development of personalized medicine approaches.
Collapse
Affiliation(s)
- Anna Chiara Siciliano
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Stefania Forciniti
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Valentina Onesto
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Helena Iuele
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Donatella Delle Cave
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, National Research Council (Cnr-IGB), Naples, 80131, Italy
| | - Federica Carnevali
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Mathematics and Physics "Ennio De Giorgi", University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- Department of Experimental Medicine, University of Salento, c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Enza Lonardo
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, National Research Council (Cnr-IGB), Naples, 80131, Italy
| | - Loretta L Del Mercato
- Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| |
Collapse
|
38
|
Xue Y, Zhang Y, Su Y, Zhao J, Yu D, Jo Y, Joo J, Lee HJ, Ryu D, Wei S. The implicated role of GDF15 in gastrointestinal cancer. Eur J Clin Invest 2024; 54:e14290. [PMID: 39044314 DOI: 10.1111/eci.14290] [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: 04/26/2024] [Accepted: 07/03/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Growth differentiation factor 15 (GDF15), a stress-responsive cytokine from transforming growth factor superfamily, is highly expressed in mammalian tissues, including pancreas, stomach and intestine under pathological conditions. In particular, elevated levels of GDF15 might play an important role in the development and progression of various gastrointestinal cancers (GCs), suggesting its potential as a promising target for disease prediction and treatment. METHODS In this review, systematic reviews addressing the role of GDF15 in GCs were updated, along with the latest clinical trials focussing on the GDF15-associated digestive malignancies. RESULTS The multiple cellular pathways through which GDF15 is involved in the regulation of physiological and pathological conditions were first summarized. Then, GDF15 was also established as a valuable clinical index, functioning as a predictive marker in diverse GCs. Notably, latest clinical treatments targeting GDF15 were also highlighted, demonstrating its promising potential in mitigating and curing digestive malignancies. CONCLUSIONS This review unveils the pivotal roles of GDF15 and its potential as a promising target in the pathogenesis of GCs, which may provide insightful directions for future investigations.
Collapse
Affiliation(s)
- Yingqi Xue
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Yan Zhang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Yale Su
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jiangqi Zhao
- Department of Dermatology, The Second Hospital of Jilin University, Changchun, China
| | - Daoquan Yu
- Department of Hepatological Surgery, Shuangliao Center Hospital, Shuangliao, China
| | - Yunju Jo
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Jongkil Joo
- Department of Obstetrics and Gynecology, Pusan National University Hospital, Busan, Korea
| | - Hyun Joo Lee
- Department of Obstetrics and Gynecology, Pusan National University Hospital, Busan, Korea
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Shibo Wei
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| |
Collapse
|
39
|
Fenu G, Griñán-Lisón C, Pisano A, González-Titos A, Farace C, Fiorito G, Etzi F, Perra T, Sabalic A, Toledo B, Perán M, Solinas MG, Porcu A, Marchal JA, Madeddu R. Unveiling the microRNA landscape in pancreatic ductal adenocarcinoma patients and cancer cell models. BMC Cancer 2024; 24:1308. [PMID: 39448959 PMCID: PMC11515555 DOI: 10.1186/s12885-024-13007-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge due to late-stage diagnoses resulting from nonspecific early symptoms and the absence of early diagnostic biomarkers. MicroRNAs (miRNAs) play a crucial role in regulating diverse biological processes, and their abnormal expression is observed in various diseases, including cancer. Cancer stem cells (CSCs) are thought to act as a driving force in PDAC spread and recurrence. In pursuing the goal of unravelling the complexities of PDAC and its underlying molecular mechanisms, our study aimed to identify PDAC-associated miRNAs and relate them to disease progression, focusing on their involvement in various PDAC stages in patients and in reliable in vitro models, including pancreatic CSC (PaCSC) models. METHODS The miRNA profiling datasets of serum and solid biopsies of PDAC patients deposited in GEO DataSets were analyzed by REML-based meta-analysis. The panel was then investigated by Real Time PCR in serum and solid biopsies of 37 PDAC patients enrolled in the study, as well as on BxPC-3 and AsPC-1 PDAC cell lines. We extended our focus towards a possible role of PDAC-associated miRNAs in the CSC phenotype, by inducing CSC-enriched pancreatospheres from BxPC-3 and AsPC-1 PDAC cell lines and performed differential miRNA expression analysis between PaCSCs and monolayer-grown PDAC cell lines. RESULTS Meta-analysis showed differentially expressed miRNAs in blood samples and cancerous tissues of PDAC patients, allowing the identification of a panel of 9 PDAC-associated miRNAs. The results emerging from our patients fully confirmed the meta-analysis for the majority of miRNAs under investigation. In vitro tasks confirmed the aberrant expression of the panel of PDAC-associated miRNAs, with a dramatic dysregulation in PaCSC models. Notably, PaCSCs have shown significant overexpression of miR-4486, miR-216a-5p, and miR-216b-5p compared to PDAC cell lines, suggesting the recruitment of such miRNAs in stemness-related molecular mechanisms. Globally, our results showed a dual behaviour of miR-216a-5p and miR-216b-5p in PDAC while miR-4486, miR-361-3p, miR-125a-5p, miR-320d expression changes during the disease suggest they could promote PDAC initiation and progression. CONCLUSIONS This study contributed to an enhanced comprehension of the role of miRNAs in the development and progression of PDAC, shedding new light on the miRNA landscape in PDAC and its intricate interplay with CSCs, and providing specific insights useful in the development of miRNA-based diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Grazia Fenu
- Department of Biomedical Science, University of Sassari, Sassari, 07100, Italy
| | - Carmen Griñán-Lisón
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, 18016, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, University of Granada, Granada, 18071, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18016, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, 18071, Spain
| | - Andrea Pisano
- Department of Biomedical Science, University of Sassari, Sassari, 07100, Italy
| | - Aitor González-Titos
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, 18016, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, University of Granada, Granada, 18071, Spain
| | - Cristiano Farace
- Department of Biomedical Science, University of Sassari, Sassari, 07100, Italy.
- National Institute of Biostructures and Biosystems, Rome, 00136, Italy.
| | - Giovanni Fiorito
- Clinical Bioinformatics Unit, IRCSS Istituto Giannina Gaslini, Genoa, 16147, Italy
| | - Federica Etzi
- Department of Biomedical Science, University of Sassari, Sassari, 07100, Italy
| | - Teresa Perra
- Department of Medicine, Surgery and Pharmacy - Unit of General Surgery, University of Sassari, Sassari, 07100, Italy
| | - Angela Sabalic
- Department of Biomedical Science, University of Sassari, Sassari, 07100, Italy
| | - Belén Toledo
- Instituto de Investigación Biosanitaria Ibs.GRANADA, University of Granada, Granada, 18071, Spain
- Department of Health Sciences, University of Jaén, Jaén, 23071, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Jaén, 23071, Spain
| | | | - Alberto Porcu
- Department of Medicine, Surgery and Pharmacy - Unit of General Surgery, University of Sassari, Sassari, 07100, Italy
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, 18016, Spain.
- Instituto de Investigación Biosanitaria Ibs.GRANADA, University of Granada, Granada, 18071, Spain.
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18016, Spain.
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18016, Spain.
| | - Roberto Madeddu
- Department of Biomedical Science, University of Sassari, Sassari, 07100, Italy
- National Institute of Biostructures and Biosystems, Rome, 00136, Italy
| |
Collapse
|
40
|
Pączek S, Zajkowska M, Mroczko B. Pigment Epithelial-Derived Factor in Pancreatic and Liver Cancers-From Inflammation to Cancer. Biomedicines 2024; 12:2260. [PMID: 39457573 PMCID: PMC11504982 DOI: 10.3390/biomedicines12102260] [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: 08/30/2024] [Revised: 09/28/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Gastrointestinal (GI) cancers are among the leading causes of mortality worldwide. Despite the emergence of new possibilities that offer hope regarding the successful treatment of these cancers, they still represent a significant global health burden. These cancers can arise from various cell types within the gastrointestinal tract and may exhibit different characteristics, behaviors, and treatment approaches. Both the prognosis and the outcomes of GI treatment remain problematic because these tumors are primarily diagnosed in advanced clinical stages. Current biomarkers exhibit limited sensitivity and specificity. Therefore, when developing strategies for the diagnosis and treatment of GI cancers, it is of fundamental importance to discover new biomarkers capable of addressing the challenges of early-stage diagnosis and the presence of lymph node metastases. Pigment epithelial-derived factor (PEDF) has garnered interest due to its inhibitory effects on the migration and proliferation of cancer cells. This protein has been suggested to be involved in various inflammation-related diseases, including cancer, through various mechanisms. It was also observed that reducing the level of PEDF is sufficient to trigger an inflammatory response. This suggests that PEDF is an endogenous anti-inflammatory factor. Overall, PEDF is a versatile protein with diverse biological functions that span across different tissues and organ systems. Its multifaceted activities make it an intriguing target for therapeutic interventions in various diseases, including cancer, neurodegeneration, and metabolic disorders. This review, for the first time, summarizes the role of PEDF in the pathogenesis of selected GI cancers and its potential utility in early diagnosis, prognosis, and therapeutic strategies for this malignancy.
Collapse
Affiliation(s)
- Sara Pączek
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland; (S.P.); (B.M.)
| | - Monika Zajkowska
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland; (S.P.); (B.M.)
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15 A, Waszyngtona St., 15-269 Białystok, Poland
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland; (S.P.); (B.M.)
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15 A, Waszyngtona St., 15-269 Białystok, Poland
- Department of Biochemical Diagnostics, Medical University of Białystok, 15-089 Białystok, Poland
| |
Collapse
|
41
|
Ahmed TM, Chu LC, Javed AA, Yasrab M, Blanco A, Hruban RH, Fishman EK, Kawamoto S. Hidden in plain sight: commonly missed early signs of pancreatic cancer on CT. Abdom Radiol (NY) 2024; 49:3599-3614. [PMID: 38782784 DOI: 10.1007/s00261-024-04334-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 05/25/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has poor prognosis mostly due to the advanced stage at which disease is diagnosed. Early detection of disease at a resectable stage is, therefore, critical for improving outcomes of patients. Prior studies have demonstrated that pancreatic abnormalities may be detected on CT in up to 38% of CT studies 5 years before clinical diagnosis of PDAC. In this review, we highlight commonly missed signs of early PDAC on CT. Broadly, these commonly missed signs consist of small isoattenuating PDAC without contour deformity, isolated pancreatic duct dilatation and cutoff, focal pancreatic enhancement and focal parenchymal atrophy, pancreatitis with underlying PDAC, and vascular encasement. Through providing commentary on demonstrative examples of these signs, we demonstrate how to reduce the risk of missing or misinterpreting radiological features of early PDAC.
Collapse
Affiliation(s)
- Taha M Ahmed
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC 3140E, 601 N Caroline St, Baltimore, MD, 21287, USA
| | - Linda C Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC 3140E, 601 N Caroline St, Baltimore, MD, 21287, USA
| | - Ammar A Javed
- Department of Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Mohammad Yasrab
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC 3140E, 601 N Caroline St, Baltimore, MD, 21287, USA
| | - Alejandra Blanco
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC 3140E, 601 N Caroline St, Baltimore, MD, 21287, USA
| | - Ralph H Hruban
- Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC 3140E, 601 N Caroline St, Baltimore, MD, 21287, USA
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC 3140E, 601 N Caroline St, Baltimore, MD, 21287, USA.
| |
Collapse
|
42
|
Ahmadi Jazi S, Tajik F, Rezagholizadeh F, Taha SR, Shariat Zadeh M, Bouzari B, Madjd Z. Higher Expression of Talin-1 is Associated With Less Aggressive Tumor Behavior in Pancreatic Cancer. Appl Immunohistochem Mol Morphol 2024; 32:425-435. [PMID: 39258796 DOI: 10.1097/pai.0000000000001220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 07/16/2024] [Indexed: 09/12/2024]
Abstract
Talin-1 is one of the major scaffold proteins in focal adhesions playing a vital role in cell migration, metastasis, and cancer progression. Although studies regarding the importance of Talin-1 in cancer have rapidly developed, its prognostic and diagnostic value still remain unsatisfying in pancreatic cancer (PC). Therefore, the present study aims to investigate the expression, clinical significance, as well as the prognostic and diagnostic value of Talin-1 in different types of PC. Bioinformatic analysis was applied to determine the clinical importance and biological role of Talin-1 expression in PC tumors and the normal adjacent samples. The expression patterns, clinical significance, prognosis, and diagnosis value of Talin-1 were evaluated in tissue microarrays (TMAs) of 190 PC samples including 170 pancreatic ductal adenocarcinoma (PDAC), and 20 pancreatic neuroendocrine tumors (PNET), along with 24 adjacent normal tissues using immunohistochemistry (IHC). The results indicated that the expression of Talin-1 was upregulated in tumor cells compared with adjacent normal tissues. A statistically significant association was observed between the higher cytoplasmic expression of Talin-1 and lower histologic grade ( P <0.001) in PDAC samples. Further, our findings indicated an inverse significant correlation between cytoplasmic expression of Talin-1 and recurrence ( P =0.014) in PNET samples. No significant association was observed between the cytoplasmic expression of Talin-1 and survival outcomes as well as diagnostic accuracy. In conclusion, our observations demonstrated that a higher cytoplasmic level of Talin-1 protein was significantly associated with less aggressive tumor behaviors in PC samples. Nevertheless, further investigations are required to explore the prognostic plus diagnostic value, and mechanism of action of Talin-1 in pancreatic cancer.
Collapse
Affiliation(s)
- Samira Ahmadi Jazi
- Department of Pathology, School of Medicine, Iran University of Medical Sciences
| | - Fatemeh Tajik
- Oncopathology Research Center, Iran University of Medical Sciences
- Department of Surgery, University of California, Irvine Medical Center, Orange, CA
| | - Fereshteh Rezagholizadeh
- Oncopathology Research Center, Iran University of Medical Sciences
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Taha
- Oncopathology Research Center, Iran University of Medical Sciences
| | | | - Behnaz Bouzari
- Department of Pathology, School of Medicine, Iran University of Medical Sciences
| | - Zahra Madjd
- Department of Pathology, School of Medicine, Iran University of Medical Sciences
- Oncopathology Research Center, Iran University of Medical Sciences
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Zhang G, Li Y, Sun Y. Cruciferous vegetables intake reduces pancreatic cancer risk: an updated systematic review with meta-analysis. Eur J Nutr 2024; 63:2421-2435. [PMID: 39078523 DOI: 10.1007/s00394-024-03472-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 07/17/2024] [Indexed: 07/31/2024]
Abstract
PURPOSE The escalating disease burden associated with pancreatic cancer has led to its inclusion as a target of public health efforts. The relationship between the consumption of cruciferous vegetables and the incidence of pancreatic cancer has generated conflicting findings in various epidemiological studies. METHODS Eligible studies were cohort or case-control studies reporting on the association between consumption of cruciferous vegetables and pancreatic cancer. PubMed, Cochrane, and Embase were systematically searched for potential studies before 27 July2022. These databases were finally searched again on 1 February 2023. The meta-analysis was conducted using random-effects models, and Stata 17 was employed for the statistical analyses. RESULTS Sixteen studies, encompassing 1,135,281 participants from 1989 to 2021, met the inclusion criteria. An inverse association of statistical significance was observed (RR 0.83, 95% CI 0.72-0.96). The sensitivity analysis indicates that this result is robust. We conducted subgroup analyses based on region, gender, study design, quality, exclusivity to endocrine tumors, adjustment for smoking, alcohol consumption, diabetes, and BMI. CONCLUSION This review provided support for the inclusion of cruciferous vegetables in the diet as a cost-effective and readily available prevention.
Collapse
Affiliation(s)
- Genyuan Zhang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110022, China
- The First Clinical College, China Medical University, Shenyang, Liaoning Province, 110022, China
| | - Yunjia Li
- The First Clinical College, China Medical University, Shenyang, Liaoning Province, 110022, China
| | - Yan Sun
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110022, China.
| |
Collapse
|
44
|
Wang F, Yan X, Peng X, Liu D, Bu W, Kang F, Song J, Wang Q. CircRNA PGAM1 Promotes the Migration and Invasion of Pancreatic Adenocarcinoma Cells by Activating the AKT/mTOR Signaling Pathway. Mol Biotechnol 2024; 66:2341-2348. [PMID: 37702882 DOI: 10.1007/s12033-023-00865-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 08/17/2023] [Indexed: 09/14/2023]
Abstract
Pancreatic adenocarcinoma (PAAD) is a lethal malignancy of the gastrointestinal tract. Circular RNA, an endogenous noncoding RNA, is considered a new regulatory molecule in tumorigenesis and development. Here, we aimed to investigate the role of circPGAM1 in PAAD. The PAAD cell line HPAC was transfected with OE-circPGAM1 to overexpress circPGAM1 and treated with AZD5363 to inhibit the AKT/mTOR pathway. Simultaneously, another PAAD cell line BxPC-3 was transfected with sh-circPGAM1 to silence circPGAM1. The GEPIA database was used to determine the expression of circPGAM1 in PAAD and its association with overall and disease-free survival. CircPGAM1 expression levels were determined in cell lines using reverse transcription-quantitative PCR. The cell counting kit-8, wound healing, and transwell assays were performed to determine cell migration and invasion. The protein expression levels of phosphorylated AKT and mTOR were determined using western blotting. CircPGAM1 was overexpressed in PAAD and related to poor prognosis. Silencing circPGAM1 inhibited migration and invasion of BxPC-3 cells, and overexpression of circPGAM1 showed the opposite effects. Overall, circPGAM1 promoted the migration and invasion of PAAD cells through the AKT/mTOR axis.
Collapse
Affiliation(s)
- Feng Wang
- Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Xingqing District, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Xiaogang Yan
- Ningxia Yinchuan First People's Hospital, Yinchuan, China
| | - Xi Peng
- Ningxia Medical University, Yinchuan, China
| | - Di Liu
- Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Xingqing District, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Wenping Bu
- Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Xingqing District, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Fuping Kang
- Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Xingqing District, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Jianjun Song
- Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Xingqing District, Yinchuan, 750004, Ningxia Hui Autonomous Region, China
| | - Qi Wang
- Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 Shengli Street, Xingqing District, Yinchuan, 750004, Ningxia Hui Autonomous Region, China.
| |
Collapse
|
45
|
Aparicio-Lopez CB, Timmerman S, Lorino G, Rogers T, Pyle M, Shrestha TB, Basel MT. Thermosensitive Liposomes for Gemcitabine Delivery to Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:3048. [PMID: 39272906 PMCID: PMC11394165 DOI: 10.3390/cancers16173048] [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: 07/26/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Treatment of pancreatic ductal adenocarcinoma with gemcitabine is limited by an increased desmoplasia, poor vascularization, and short plasma half-life. Heat-sensitive liposomes modified by polyethylene glycol (PEG; PEGylated liposomes) can increase plasma stability, reduce clearance, and decrease side effects. Nevertheless, translation of heat-sensitive liposomes to the clinic has been hindered by the low loading efficiency of gemcitabine and by the difficulty of inducing hyperthermia in vivo. This study was designed to investigate the effect of phospholipid content on the stability of liposomes at 37 °C and their release under hyperthermia conditions; this was accomplished by employing a two-stage heating approach. First the liposomes were heated at a fast rate, then they were transferred to a holding bath. Thermosensitive liposomes formulated with DPPC: DSPC: PEG2k (80:15:5, mole%) exhibited minimal release of carboxyfluorescein at 37 °C over 30 min, indicating stability under physiological conditions. However, upon exposure to hyperthermic conditions (43 °C and 45 °C), these liposomes demonstrated a rapid and significant release of their encapsulated content. The encapsulation efficiency for gemcitabine was calculated at 16.9%. Additionally, fluorescent analysis during the removal of unencapsulated gemcitabine revealed an increase in pH. In vitro tests with BxPC3 and KPC cell models showed that these thermosensitive liposomes induced a heat-dependent cytotoxic effect comparable to free gemcitabine at temperatures above 41 °C. This study highlights the effectiveness of the heating mechanism and cell models in understanding the current challenges in developing gemcitabine-loaded heat-sensitive liposomes.
Collapse
Affiliation(s)
- Cesar B Aparicio-Lopez
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Sarah Timmerman
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Gabriella Lorino
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Tatiana Rogers
- Department of Electrical and Computer Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - Marla Pyle
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Tej B Shrestha
- Nanotechnology Innovation Center of Kansas State (NICKS), Kansas State University, Manhattan, KS 66506, USA
| | - Matthew T Basel
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
46
|
Xu D, Jia M, Yang F, Zhang X, Jiang K. Analyzing the role of TM4SF1 expression in pancreatic adenocarcinoma: understanding prognostic implications and therapeutic opportunities. J Gastrointest Oncol 2024; 15:1760-1776. [PMID: 39279979 PMCID: PMC11399867 DOI: 10.21037/jgo-24-564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 08/17/2024] [Indexed: 09/18/2024] Open
Abstract
Background Pancreatic adenocarcinoma (PAAD) is a highly lethal malignancy characterized by aggressive growth and poor prognosis. Understanding the molecular mechanisms underlying PAAD is crucial for developing effective therapies. This study aimed to explore the role of TM4SF1 and other key genes in PAAD progression, their prognostic implications, and therapeutic opportunities. Methods Differential gene expression analysis was performed using PAAD and normal tissue samples to identify upregulated genes, with TM4SF1 emerging as significantly elevated in PAAD. Functional enrichment analysis elucidated associated signaling pathways. A prognostic model comprising BPIFB4, PLEKHN1, CPTP, DVL1, and DDR1 was developed using least absolute shrinkage and selection operator (LASSO) regression and validated in an independent cohort. Genetic mutation analysis provided insights into the functional significance of identified genes. Pharmacogenomic analysis examined associations between gene expression and drug sensitivity. Experimental validation included quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blot analyses to confirm gene expression patterns and protein levels. Results Lower TM4SF1 expression correlated with enhanced anti-tumor immune activity in PAAD, suggesting a complex interplay between genetic expression and immune response. The prognostic model showed robust associations with patient survival outcomes, validated across diverse patient cohorts. Genetic mutation analysis highlighted potential therapeutic targets. Pharmacogenomic analysis revealed correlations between gene expression profiles and drug responsiveness, suggesting personalized treatment strategies. Experimental validation confirmed elevated TM4SF1 levels in tumor tissues and demonstrated its role in promoting cancer cell proliferation and colony formation. Conclusions This study advances understanding of the molecular landscape of PAAD, emphasizing TM4SF1 as a key regulator and potential therapeutic target. The integration of genetic expression, immune response dynamics, and pharmacogenomics offers a multifaceted approach to personalized treatment strategies for PAAD, paving the way for improved patient outcomes and novel therapeutic interventions. Further research is warranted to elucidate the clinical utility of targeting TM4SF1 and other identified genes in PAAD management.
Collapse
Affiliation(s)
- Dong Xu
- Pancreas Center & Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of General Surgery, Gaochun People's Hospital, Nanjing, China
| | - Mingguang Jia
- Department of General Surgery, Zibo Municipal Hospital, Zibo, China
| | - Fei Yang
- Department of General Surgery, Gaochun People's Hospital, Nanjing, China
| | - Xiaohui Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kuirong Jiang
- Pancreas Center & Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
47
|
Cortiana V, Vallabhaneni H, Gambill J, Nadar S, Itodo K, Park CH, Leyfman Y. Advancing Pancreatic Cancer Surgical Treatments and Proposal of New Approaches. Cancers (Basel) 2024; 16:2848. [PMID: 39199619 PMCID: PMC11352325 DOI: 10.3390/cancers16162848] [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: 06/26/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Pancreatic cancer is a significant challenge in oncology due to its aggressive nature and complex management, leading to high mortality rates and a dismally low 5-year survival rate. Approximately 85% of cases manifest as adenocarcinoma, while endocrine tumors constitute less than 5%. Borderline resectable and locally advanced pancreatic cancers are particularly difficult to treat due to vascular involvement, which complicates complete resections and increases morbidity. Various therapeutic modalities aim to overcome these challenges and improve patient outcomes. Traditionally, upfront surgery was the standard for resectable tumors, with multimodal chemotherapy being central to treatment. Understanding surgical anatomy is pivotal in enhancing surgical outcomes and patient survival. Resectability challenges are several when seeking to achieve R0 resections, particularly for borderline resectable tumors. Various classification systems-the MD Anderson criteria, the NCCN criteria, the AHPA/SSAT/SSO consensus statement, and the Alliance definition-assess tumor involvement with major blood vessels, with the first of these systems being broadly accepted. Vascular staging integration is also important, with the Ishikawa staging system using preoperative imaging to assess venous involvement. Furthermore, neoadjuvant therapy enhances treatment effectiveness by addressing micro-metastatic disease early, increasing R0 resection chances, and downstaging tumors for optimal surgery. Insights from the Fox Chase Cancer Center's neoadjuvant treatment approach highlight the importance of a multidisciplinary strategy when advancing therapy and improving patient prognosis. This commentary, inspired by Dr. Sanjay S. Reddy's Keynote Conference during MedNews week, highlights current advancements and ongoing challenges in the treatment of pancreatic cancer, emphasizing the need for a comprehensive, multidisciplinary approach to improve outcomes.
Collapse
Affiliation(s)
- Viviana Cortiana
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | | | | | - Soumiya Nadar
- Tbilisi State Medical University, 0186 Tbilisi, Georgia
| | - Kennedy Itodo
- Nigerian Institute for Trypanosomiasis Research Jos, Kaduna PMB 2077, Nigeria
| | | | - Yan Leyfman
- Icahn School of Medicine at Mount Sinai South Nassau, Oceanside, NY 11572, USA;
| |
Collapse
|
48
|
Ahmad I, Jasim SA, Sharma MK, S RJ, Hjazi A, Mohammed JS, Sinha A, Zwamel AH, Hamzah HF, Mohammed BA. New paradigms to break barriers in early cancer detection for improved prognosis and treatment outcomes. J Gene Med 2024; 26:e3730. [PMID: 39152771 DOI: 10.1002/jgm.3730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 08/19/2024] Open
Abstract
The uncontrolled growth and spread of cancerous cells beyond their usual boundaries into surrounding tissues characterizes cancer. In developed countries, cancer is the leading cause of death, while in underdeveloped nations, it ranks second. Using existing cancer diagnostic tools has increased early detection rates, which is crucial for effective cancer treatment. In recent decades, there has been significant progress in cancer-specific survival rates owing to advances in cancer detection and treatment. The ability to accurately identify precursor lesions is a crucial aspect of effective cancer screening programs, as it enables early treatment initiation, leading to lower long-term incidence of invasive cancer and improved overall prognosis. However, these diagnostic methods have limitations, such as high costs and technical challenges, which can make accurate diagnosis of certain deep-seated tumors difficult. To achieve accurate cancer diagnosis and prognosis, it is essential to continue developing cutting-edge technologies in molecular biology and cancer imaging.
Collapse
Affiliation(s)
- Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-maarif, Anbar, Iraq
| | - M K Sharma
- Department of Mathematics, Chaudhary Charan Singh University, Meerut, Uttar Pradesh, India
| | - Renuka Jyothi S
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Ahmed Hussein Zwamel
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| | - Hamza Fadhel Hamzah
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | |
Collapse
|
49
|
Telisnor G, Lim A, Zhang Z, Lou X, Nassour I, Salloum RG, Rogers SC. Analysis of pancreatic cancer treatment and survival disparities in Florida throughout the Covid-19 pandemic. J Natl Med Assoc 2024; 116:328-337. [PMID: 39107147 DOI: 10.1016/j.jnma.2024.07.004] [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: 08/08/2023] [Revised: 11/27/2023] [Accepted: 07/02/2024] [Indexed: 08/09/2024]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is currently the third-leading cause of cancer-related death in the United States. African Americans (AAs) with PDAC have worse survival in comparison to other racial groups. The COVID-19 pandemic caused significant stress to the healthcare system. We aim to evaluate the pandemic's impact on already known disparities in newly diagnosed patients with PDAC in Florida. METHODS This is a retrospective analysis of newly diagnosed patients with PDAC in the OneFlorida+ Data Trust based upon date of diagnosis: Pre-pandemic (01/01/2017- 09/30/2019), Transition (10/01/2019-02/28/2020), and Pandemic (03/1/2020-10/31/2020). Primary endpoints are time to treatment initiation and rate of surgery and secondary endpoint is survival time. Disparities due to age, sex, race, and income were also evaluated. Chi-squared or Fisher's exact test when necessary, Kruskal-Wallis test, and Kaplan-Meier analysis with log-rank test were performed to compare the differences between the comparative groups for categorical, quantitative, and survival outcomes, respectively. Multivariable regression analyses were conducted to estimate the effects of cofactors. RESULTS 934 patients with a median age of 67 years were included. There were 47.8% females and 52.2% males; 19.4% AA, 70.2% Caucasian, 10.4% Other race; median income was $53,551. While we observed a significant reduction in the diagnosis rate of new PDAC cases during the pandemic, there were no significant differences in demographic distributions among the three cohorts. Time to treatment did not significantly change from the pre-pandemic to the pandemic, and no difference was observed across all demographics. Rate of surgery increased significantly from the pre-pandemic (35.8%) to the pandemic (55.6%). AAs in the pre-pandemic cohort had a significantly lower rate of surgery of 25.0% compared to 41.7% in Caucasians. AAs, patients ≥ 67 years, and income < $53,000 had significantly higher hazards to death and shorter median survival time (mST). CONCLUSIONS While no differences in time to initial treatment are observed among the newly diagnosed PDAC patients, there remain significant disparities in the rate of surgery and overall survival. Observing a significant reduction in diagnosis rate and analyzing disparities can provide insight into the effect of a resource-restricting pandemic for patients with newly diagnosed PDAC.
Collapse
Affiliation(s)
- Guettchina Telisnor
- University of Florida, College of Pharmacy, Gainesville, Florida, United States
| | - Alexander Lim
- University of Florida, Department of Medicine, Division of Hematology and Oncology, Gainesville, Florida, United States
| | - Zhongyue Zhang
- Division of Quantitative Science, University of Florida Health Cancer Center, Gainesville, Florida, United States
| | - XiangYang Lou
- University of Florida, Department of Biostatistics, Gainesville, Florida, United States
| | - Ibrahim Nassour
- University of Florida, Department of Surgery, Division of Surgical Oncology, Gainesville, Florida, United States
| | - Ramzi G Salloum
- University of Florida, Department of Health Outcomes and Biomedical Informatics, Gainesville, Florida, United States
| | - Sherise C Rogers
- University of Florida, Department of Medicine, Division of Hematology and Oncology, Gainesville, Florida, United States.
| |
Collapse
|
50
|
Liu W, Zhang B, Liu T, Jiang J, Liu Y. Artificial Intelligence in Pancreatic Image Analysis: A Review. SENSORS (BASEL, SWITZERLAND) 2024; 24:4749. [PMID: 39066145 PMCID: PMC11280964 DOI: 10.3390/s24144749] [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: 06/05/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Pancreatic cancer is a highly lethal disease with a poor prognosis. Its early diagnosis and accurate treatment mainly rely on medical imaging, so accurate medical image analysis is especially vital for pancreatic cancer patients. However, medical image analysis of pancreatic cancer is facing challenges due to ambiguous symptoms, high misdiagnosis rates, and significant financial costs. Artificial intelligence (AI) offers a promising solution by relieving medical personnel's workload, improving clinical decision-making, and reducing patient costs. This study focuses on AI applications such as segmentation, classification, object detection, and prognosis prediction across five types of medical imaging: CT, MRI, EUS, PET, and pathological images, as well as integrating these imaging modalities to boost diagnostic accuracy and treatment efficiency. In addition, this study discusses current hot topics and future directions aimed at overcoming the challenges in AI-enabled automated pancreatic cancer diagnosis algorithms.
Collapse
Affiliation(s)
- Weixuan Liu
- Sydney Smart Technology College, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China; (W.L.); (B.Z.)
| | - Bairui Zhang
- Sydney Smart Technology College, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China; (W.L.); (B.Z.)
| | - Tao Liu
- School of Mathematics and Statistics, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China;
| | - Juntao Jiang
- College of Control Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yong Liu
- College of Control Science and Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|