1
|
Casarcia N, Rogers P, Guld E, Iyer S, Li Y, Burcher JT, DeLiberto LK, Banerjee S, Bishayee A. Phytochemicals for the prevention and treatment of pancreatic cancer: Current progress and future prospects. Br J Pharmacol 2025; 182:2181-2234. [PMID: 37740585 DOI: 10.1111/bph.16249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/06/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023] Open
Abstract
Pancreatic cancer is the third leading cause of cancer-related deaths in the United States, owing to its aggressive nature and suboptimal treatment options, emphasizing the need for novel therapeutic approaches. Emerging studies have exhibited promising results regarding the therapeutic utility of plant-derived compounds (phytochemicals) in pancreatic cancer. The purpose of this review is to evaluate the potential of phytochemicals in the treatment and prevention of pancreatic cancer. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses was applied to collect articles for this review. Scholarly databases, including PubMed, Scopus and ScienceDirect, were queried for relevant studies using the following keywords: phytochemicals, phenolics, terpenoids, alkaloids, sulfur-containing compounds, in vitro, in vivo, clinical studies, pancreatic cancer, tumour, treatment and prevention. Aggregate results pooled from qualified studies indicate phytochemicals can inhibit pancreatic cancer cell growth or decrease tumour size and volume in animal models. These effects have been attributed to various mechanisms, such as increasing proapoptotic factors, decreasing antiapoptotic factors, or inducing cell death and cell cycle arrest. Notable signalling pathways modulated by phytochemicals include the rat sarcoma/mitogen activated protein kinase, wingless-related integration site/β-catenin and phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin signal transduction pathways. Clinically, phytochemicals have been found to increase survival while being well-tolerated and safe, though research is scarce. While these promising results have produced great interest in this field, further in-depth studies are required to characterize the anticancer activities of phytochemicals before they can be utilized to prevent or treat pancreatic cancer in clinical practice. LINKED ARTICLES: This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Nicolette Casarcia
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Patrick Rogers
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Emma Guld
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Samvit Iyer
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Yutong Li
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Jack T Burcher
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Lindsay K DeLiberto
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol, India
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| |
Collapse
|
2
|
Li Z, Zhang T, Yang X, Peng Y. Role of noncoding RNA and protein interaction in pancreatic cancer. Chin Med J (Engl) 2025:00029330-990000000-01514. [PMID: 40205638 DOI: 10.1097/cm9.0000000000003587] [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: 01/01/2025] [Indexed: 04/11/2025] Open
Abstract
ABSTRACT Noncoding RNAs (ncRNAs) are a class of RNA molecules with little or no protein-coding potential. Emerging evidence indicates that ncRNAs are frequently dysregulated and play pivotal roles in the pathogenesis of pancreatic cancer. Their aberrant expression can arise from chromosomal abnormalities, dysregulated transcriptional control, and epigenetic modifications. ncRNAs function as protein scaffolds or molecular decoys to modulate interactions between proteins and other biomolecules, thereby regulating gene expression and contributing to pancreatic cancer progression. In this review, we summarize the mechanisms underlying ncRNA dysregulation in pancreatic cancer, emphasize the biological significance of ncRNA-protein interactions, and highlight their clinical relevance. A deeper understanding of ncRNA-protein interactions is essential to elucidate molecular mechanisms and advance translational research in pancreatic cancer.
Collapse
Affiliation(s)
- Zhang Li
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | | | | | | |
Collapse
|
3
|
Sharma R, Kumar S, Komal K, Ghosh R, Thakur S, Pal RR, Kumar M. Comprehensive insights into pancreatic cancer treatment approaches and cutting-edge nanocarrier solutions: from pathology to nanomedicine. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04094-y. [PMID: 40202672 DOI: 10.1007/s00210-025-04094-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025]
Abstract
Pancreatic cancer is one of the most lethal malignancies worldwide. It is characterized by poor prognosis, high mortality, and recurrence rates. Various modifiable and non-modifiable risk factors are associated with pancreatic cancer incidence. Available treatments for pancreatic cancer include surgery, chemotherapy, radiotherapy, photodynamic therapy, supportive care, targeted therapy, and immunotherapy. However, the survival rates for PC are very low. Regrettably, despite efforts to enhance prognosis, the survival rate of pancreatic cancer remains relatively low. Therefore, it is essential to investigate new approaches to improve pancreatic cancer treatment. By synthesizing current knowledge and identifying existing gaps, this article provides a comprehensive overview of risk factors, pathology, conventional treatments, targeted therapies, and recent advancements in nanocarriers for its treatment, along with various clinical trials and patents that justify the safety and efficacy of innovative carriers for drug delivery systems. Ultimately, this review underscores the potential of these innovative formulations to improve outcomes and contribute significantly to the advancement of Pancreatic Cancer treatment. Together, these insights highlight nano-formulations as a promising frontier for effectively treating Pancreatic Cancer.
Collapse
Affiliation(s)
- Rohit Sharma
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Kumari Komal
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Rashmi Ghosh
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Shubham Thakur
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Ravi Raj Pal
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
4
|
Thakral F, Prasad B, Sehgal R, Gupta S, Sharma U, Singh BJ, Sharma B, Tuli HS, Haque S, Ahmad F. Role of emodin to prevent gastrointestinal cancers: recent trends and future prospective. Discov Oncol 2025; 16:468. [PMID: 40186678 PMCID: PMC11972247 DOI: 10.1007/s12672-025-02240-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/25/2025] [Indexed: 04/07/2025] Open
Abstract
Gastrointestinal malignancies are responsible for approximately 35% of all cancer-related deaths, underscoring the critical need to explore pharmacologically active molecules for chemoprevention. Emodin (1,3,8-trihydroxy-6-methylanthraquinone), a natural compound derived from traditional Chinese and Japanese medicine, has recently garnered significant attention for its potential anticancer properties. Emodin exerts its chemoprotective effects through a combination of antioxidative, anti-inflammatory, and anti-proliferative mechanisms. Research indicates that emodin inhibits cancer metastasis, disrupts cell cycle progression, and impairs cancer cell survival. These effects are mediated through the activation of the p38 MAPK/JNK1/2 signaling pathway, the upregulation of pro-apoptotic factors such as Bax/Bcl-2 and caspases, and the enhancement of reactive oxygen species (ROS) levels (Supplementary Fig. 1). To optimize emodin's therapeutic potential, it is crucial to further investigate its underlying mechanisms of action and develop advanced nano-targeted delivery systems to enhance its bioavailability. This review highlights emodin's promise as a chemopreventive agent for gastrointestinal cancers and emphasizes its potential for development into a novel clinical formulation.
Collapse
Affiliation(s)
- Falak Thakral
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, India
| | - Bhairav Prasad
- Department of Biotechnology, Chandigarh Group of Colleges, Landran, Mohali, Punjab, India
| | - Rippin Sehgal
- Department of Biotechnology, Ambala College of Engineering and Applied Research, Devsthali, Ambala, Haryana, 133101, India
| | | | - Ujjawal Sharma
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bhatinda, 151001, India
| | - Bikram Jit Singh
- Mechanical Engineering Department, MM Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, Haryana, 133207, India
| | - Bunty Sharma
- Department of Biotechnology, Graphic Era (Deemed to Be University), Dehradun, Uttarakhand, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, India
| | - Shafiul Haque
- Department of Nursing, College of Nursing and Health Sciences, Jazan University, Jazan-45142, Saudi Arabia
- School of Medicine, Universidad Espiritu Santo, Samborondon, 091952, Ecuador
| | - Faraz Ahmad
- Department of Biotechnology, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology, Vellore, 632014, India.
| |
Collapse
|
5
|
Alloubani A, Abadalhaq B, Alshami A, Fakhory D, Abdalghani F, Almasri M, Alkouz M. Exploring the prognostic significance of vitamin D deficiency in pancreatic cancer: Disease progression and survival outcomes. Cancer Treat Res Commun 2025; 43:100917. [PMID: 40222311 DOI: 10.1016/j.ctarc.2025.100917] [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: 07/25/2024] [Revised: 03/27/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Pancreatic cancer remains one of the most aggressive malignancies with limited treatment options and poor survival rates. Vitamin D deficiency has been suggested to influence cancer progression and survival outcomes in various malignancies. AIM This study aimed to investigate the association between Vitamin D deficiency and disease progression as well as survival in patients diagnosed with pancreatic cancer. METHODS A retrospective cohort study was conducted, including 151 patients diagnosed with pancreatic cancer between 2012 and 2022. Serum Vitamin D levels at the time of diagnosis were measured. Disease progression was evaluated through radiological assessments and clinical reports. Survival outcomes, including overall survival (OS) and progression-free survival (PFS), were analyzed using Kaplan-Meier survival curves and Cox proportional hazards regression models. RESULTS Of the 151 patients, 84 (56 %) were identified as Vitamin D deficient at the time of diagnosis. The deficient group exhibited a significantly higher frequency of advanced-stage disease (stages III and IV) compared to the non-deficient group (p < 0.05). During the follow-up period, 66 (78.6 %) of Vitamin d-deficient patients and 56 (84.8 %) of non-deficient patients experienced disease progression (p = 0.51). Moreover, Kaplan-Meier analysis showed a non-significant trend toward shorter median PFS (8.95 months vs. 9.27 months, p = 0.51) and OS (17.64 months vs. 19.05 months, p = 0.616) in the Vitamin d-deficient group. CONCLUSION Vitamin D deficiency is prevalent among patients with pancreatic cancer and appears to be associated with more advanced disease at diagnosis. Although a trend toward poorer survival outcomes was observed, the association between Vitamin D deficiency and OS/PFS did not reach statistical significance. Additional prospective studies are needed to confirm these findings and explore potential benefits of Vitamin D supplementation in pancreatic cancer management.
Collapse
|
6
|
Zhou K, Liu Y, Tang C, Zhu H. Pancreatic Cancer: Pathogenesis and Clinical Studies. MedComm (Beijing) 2025; 6:e70162. [PMID: 40182139 PMCID: PMC11965705 DOI: 10.1002/mco2.70162] [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: 06/24/2024] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy, with pancreatic ductal adenocarcinoma (PDAC) being the most common and aggressive subtype, characterized by late diagnosis, aggressive progression, and resistance to conventional therapies. Despite advances in understanding its pathogenesis, including the identification of common genetic mutations (e.g., KRAS, TP53, CDKN2A, SMAD4) and dysregulated signaling pathways (e.g., KRAS-MAPK, PI3K-AKT, and TGF-β pathways), effective therapeutic strategies remain limited. Current treatment modalities including chemotherapy, targeted therapy, immunotherapy, radiotherapy, and emerging therapies such as antibody-drug conjugates (ADCs), chimeric antigen receptor T (CAR-T) cells, oncolytic viruses (OVs), cancer vaccines, and bispecific antibodies (BsAbs), face significant challenges. This review comprehensively summarizes these treatment approaches, emphasizing their mechanisms, limitations, and potential solutions, to overcome these bottlenecks. By integrating recent advancements and outlining critical challenges, this review aims to provide insights into future directions and guide the development of more effective treatment strategies for PC, with a specific focus on PDAC. Our work underscores the urgency of addressing the unmet needs in PDAC therapy and highlights promising areas for innovation in this field.
Collapse
Affiliation(s)
- Kexun Zhou
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Yingping Liu
- Department of RadiotherapyCancer HospitalChinese Academy of Medical SciencesBeijingChina
| | - Chuanyun Tang
- The First Clinical Medical College of Nanchang UniversityNanchang UniversityNanchangChina
| | - Hong Zhu
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengduChina
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
7
|
Powrózek T, Otieno MO, Maffeo D, Frullanti E, Martinez-Useros J. Blood circulating miRNAs as pancreatic cancer biomarkers: An evidence from pooled analysis and bioinformatics study. Int J Biol Macromol 2025:142469. [PMID: 40180095 DOI: 10.1016/j.ijbiomac.2025.142469] [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: 08/08/2024] [Revised: 03/09/2025] [Accepted: 03/22/2025] [Indexed: 04/05/2025]
Abstract
Pancreatic cancer (PC) is one of the deadliest cancers, characterized by a poor prognosis. Currently, there are no screening programs for the early detection of PC, and existing diagnostic methods are primarily limited to high-risk individuals. Biomarkers such as CA19-9 have not significantly improved early diagnosis, making the identification of new potential biomarkers crucial for routine clinical practice. Among the candidate biomarkers, miRNAs have been most extensively studied due to their role in regulating gene expression (either as oncomiRs or tumor suppressor miRNAs) and their potential for minimally invasive analysis through liquid biopsy techniques. This review aims to summarize the current literature on blood-circulating miRNAs and their diagnostic value in PC detection, considering the context of CA19-9 and benign pancreatic diseases. The data from the collected studies were curated through both statistical and bioinformatics analyses to identify the most promising miRNAs with optimal diagnostic accuracy for PC detection and to assess their role in the molecular processes leading to tumor development.
Collapse
Affiliation(s)
- Tomasz Powrózek
- Department of Human Physiology, Medical University of Lublin, Lublin, Poland.
| | - Michael Ochieng' Otieno
- Translational Oncology Division, Oncohealth Institute, Fundacion Jiménez Díaz University Hospital, Madrid, Spain
| | - Debora Maffeo
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Cancer Genomics and Systems Biology Lab, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elisa Frullanti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Cancer Genomics and Systems Biology Lab, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Javier Martinez-Useros
- Translational Oncology Division, Oncohealth Institute, Fundacion Jiménez Díaz University Hospital, Madrid, Spain; Area of Physiology, Department of Basic Health Sciences, Faculty of Health Sciences, Rey Juan Carlos University, Madrid, Spain
| |
Collapse
|
8
|
Huang C, Shen Y, Galgano SJ, Goenka AH, Hecht EM, Kambadakone A, Wang ZJ, Chu LC. Advancements in early detection of pancreatic cancer: the role of artificial intelligence and novel imaging techniques. Abdom Radiol (NY) 2025; 50:1731-1743. [PMID: 39467913 DOI: 10.1007/s00261-024-04644-7] [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: 08/15/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024]
Abstract
Early detection is crucial for improving survival rates of pancreatic ductal adenocarcinoma (PDA), yet current diagnostic methods can often fail at this stage. Recently, there has been significant interest in improving risk stratification and developing imaging biomarkers, through novel imaging techniques, and most notably, artificial intelligence (AI) technology. This review provides an overview of these advancements, with a focus on deep learning methods for early detection of PDA.
Collapse
Affiliation(s)
| | - Yiqiu Shen
- New York University Langone Health, New York, USA
| | | | | | | | | | - Zhen Jane Wang
- University of California, San Francisco, San Francisco, USA
| | - Linda C Chu
- Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
9
|
Santucci C, Mignozzi S, Alicandro G, Pizzato M, Malvezzi M, Negri E, Jha P, La Vecchia C. Trends in cancer mortality under age 50 in 15 upper-middle and high-income countries. J Natl Cancer Inst 2025; 117:747-760. [PMID: 39576674 PMCID: PMC11972687 DOI: 10.1093/jnci/djae288] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/25/2024] [Accepted: 11/06/2024] [Indexed: 04/08/2025] Open
Abstract
BACKGROUND Rising cancer incidence, particularly for colorectal cancer, has been reported in young adults. This study examined whether this is related to an increase in mortality. METHODS We analyzed World Health Organization mortality data among young adults aged 25-49 years in 15 most populous upper-middle and high-income countries from 1990 to 2021 with reliable data. Midyear populations were retrieved from the United Nations for the American Countries and from the World Health Organization for the other countries. We compared age-standardized mortality rates in 2019-2021 with those in 2009-2011 and performed joinpoint regression analysis for all cancers and selected most common cancer sites: colorectum, pancreas, lung, and breast. RESULTS In 2019-2021, the highest age-standardized mortality rates (per 100 000) were in Romanian males (38.6) and Argentinian females (45.9), while the lowest ones were in Japanese males (16.3) and females (22.7). Age-standardized mortality rates for colorectal cancers increased in 2019-2021 compared with 2009-2011 in 9 countries among men and in 7 countries among women. The highest increases were in the United Kingdom (males: +26.1%; females: +33.7%), Canada (males: +25.3%), and Mexico (males: +33.5%; females: +29.7%). Long-term analysis over the last 3 decades showed declining trends in total cancer mortality in the majority of countries, in lung cancer mortality across all countries, and in breast cancer in all countries except in Latin America. CONCLUSIONS Although mortality from common cancers has generally decreased over the past 3 decades, mortality from colorectal cancer has increased in some countries. This highlights the need to control the obesity epidemic and implement targeted surveillance strategies in young populations.
Collapse
Affiliation(s)
- Claudia Santucci
- Department of Clinical Sciences and Community Health, Department of Excellence 2023-2027, University of Milan, 20133 Milan, Italy
| | - Silvia Mignozzi
- Department of Clinical Sciences and Community Health, Department of Excellence 2023-2027, University of Milan, 20133 Milan, Italy
| | - Gianfranco Alicandro
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
- Mother and Child Department, Cystic Fibrosis Centre, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Margherita Pizzato
- Department of Clinical Sciences and Community Health, Department of Excellence 2023-2027, University of Milan, 20133 Milan, Italy
| | - Matteo Malvezzi
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Eva Negri
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Prabhat Jha
- Centre for Global Health Research, Unity Health Toronto, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5B 1W8, Canada
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, Department of Excellence 2023-2027, University of Milan, 20133 Milan, Italy
| |
Collapse
|
10
|
Sharma R, Ghosh R, Kumar S, Komal K, Kumar M. Decoding pancreatic cancer: key risk factors across genetics and lifestyle. Expert Rev Mol Diagn 2025; 25:95-99. [PMID: 39918844 DOI: 10.1080/14737159.2025.2464563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/05/2025] [Indexed: 02/09/2025]
Affiliation(s)
- Rohit Sharma
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Rashmi Ghosh
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Kumari Komal
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| |
Collapse
|
11
|
Zhao X, Wu G, Tao X, Dong D, Liu J. Targeted mitochondrial therapy for pancreatic cancer. Transl Oncol 2025; 54:102340. [PMID: 40048984 PMCID: PMC11928980 DOI: 10.1016/j.tranon.2025.102340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/05/2025] [Accepted: 02/27/2025] [Indexed: 03/18/2025] Open
Abstract
Pancreatic cancer (PC) is a highly invasive tumor characterized by delayed diagnosis, rapid progress, and resistance to chemotherapy. Mitochondria, as the "power chamber" of cells, not only play a central role in energy metabolism but also participate in the production of reactive oxygen species (ROS), calcium signaling, regulation, and differentiation of the cell cycle. The abnormal activity of mitochondria is closely related to the development of PC. In this paper, we discussed the key role of mitochondria in PC, including mitochondrial DNA, mitochondrial biogenesis, mitochondrial dynamics, metabolic regulation, ROS generation, and mitochondrial-dependent apoptosis. We elaborated on the importance of these mitochondrial mechanisms in the development of PC and emphasized the potential of targeted mitochondrial therapy strategies for these mechanisms in the treatment of PC. In addition, this article also reviews the latest developments in innovative drug carriers such as cell-penetrating peptides, nucleic acid aptamers, and nanomaterials, which can achieve precise localization of mitochondria and drug delivery. Therefore, this article comprehensively analyzed the important role of mitochondria in the treatment of PC and clarified the effectiveness and necessity of targeting mitochondria in the treatment of PC.
Collapse
Affiliation(s)
- Xinya Zhao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Guoyu Wu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Jing Liu
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
12
|
Zaidh SM, Vengateswaran HT, Habeeb M, Aher KB, Bhavar GB, Irfan N, Lakshmi KNVC. Network pharmacology and AI in cancer research uncovering biomarkers and therapeutic targets for RALGDS mutations. Sci Rep 2025; 15:10938. [PMID: 40157967 PMCID: PMC11954960 DOI: 10.1038/s41598-025-91568-x] [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: 09/13/2024] [Accepted: 02/21/2025] [Indexed: 04/01/2025] Open
Abstract
The lack of target therapies is accountable for the higher mortality of various types of cancer. To address this issue, we selected a target mutated Kirsten rat sarcoma virus oncogene homologue, which plays a significant role in various cancers. Our study aims to identify selective biomarkers and develop diagnostic and therapeutic strategies for KRAS-associated genes using artificial intelligence. Initially, Genomic data, cancer epidemiology, proteomics network interactions, and omics enrichment were analyzed. Structured E-pharmacophore model aided in capturing the binding cavity using eraser algorithms and fabricating a new selective lead compound for the KRSA. The selective molecule was abridged inside the binding cavity and stability was validated through 100 ns molecular dynamics simulations. Epidemiological-neural network studies indicated KRAS mutations leads 40 types of cancer, exclusively pancreatic and colorectal cancers, with diploid and missense mutations as primary factors. Pathway analysis highlighted the involvement of the MAPK and RAS signaling pathways in cancer development and proteomics analysis identified RALGDS as a key protein. Protein-based pharmacophore analysis mapped the biologically active features such as donor, acceptor and aromatic ring with the designed ligands. The results of interaction interpretation illustrate that the amino acid Tyr566 formed an H-bond interaction with the amine group of the octyl ring system and 20 amino acids crafted to properly orient the molecule to fit inside the polar cavity of KRAS protein. The MMGBSA score of - 53.33 kcal/mol conformed to the well-configured binding with KRSA and realistic model simulation exposed the π-π, π-cationic and hydrophobic interactions stabilised the molecule inside the KRSA protein throughout 100 ns simulation. The study demonstrates the vitality of AI and network pharmacology to identify potential-target biomarkers for KRAS-associated genes, paving the way for improved cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- S Mohammed Zaidh
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India
- D3 Drug Tech Lab Pvt Ltd, Chennai, 600048, India
| | | | - Mohammad Habeeb
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India
| | - Kiran Balasaheb Aher
- Department of Pharmaceutical Quality Assurance, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra, 424001, India
| | - Girija Balasaheb Bhavar
- Department of Pharmaceutical Chemistry, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra, 424001, India
| | - N Irfan
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India.
| | - K N V Chenchu Lakshmi
- Department of Pharmaceutical Chemistry, KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Green Fields, Vaddeswaram, A.P, 522302, India
| |
Collapse
|
13
|
Lo EKW, Idrizi A, Tryggvadottir R, Zhou W, Hou W, Ji H, Cahan P, Feinberg AP. DNA methylation memory of pancreatic acinar-ductal metaplasia transition state altering Kras-downstream PI3K and Rho GTPase signaling in the absence of Kras mutation. Genome Med 2025; 17:32. [PMID: 40156071 PMCID: PMC11951614 DOI: 10.1186/s13073-025-01452-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND A critical area of recent cancer research is the emergence of transition states between normal and cancer that exhibit increased cell plasticity which underlies tumor cell heterogeneity. Pancreatic ductal adenocarcinoma (PDAC) can arise from the combination of a transition state termed acinar-to-ductal metaplasia (ADM) and a gain-of-function mutation in the proto-oncogene KRAS. During ADM, digestive enzyme-producing acinar cells acquire a transient ductal epithelium-like phenotype while maintaining their geographical acinar organization. One route of ADM initiation is the overexpression of the Krüppel-like factor 4 gene (KLF4) in the absence of oncogenic driver mutations. Here, we asked to what extent cells acquire and retain an epigenetic memory of the ADM transition state in the absence of oncogene mutation. METHODS We profiled the DNA methylome and transcriptome of KLF4-induced ADM in transgenic mice at various timepoints during and after recovery from ADM. We validated the identified DNA methylation and transcriptomic signatures in the widely used caerulein model of inducible pancreatitis. RESULTS We identified differential DNA methylation at Kras-downstream PI3K and Rho/Rac/Cdc42 GTPase pathway genes during ADM, as well as a corresponding gene expression increase in these pathways. Importantly, differential methylation persisted after gene expression returned to normal. Caerulein exposure, which induces widespread digestive system changes in addition to ADM, showed similar changes in DNA methylation in ADM cells. Regions of differential methylation were enriched for motifs of KLF and AP-1 family transcription factors, as were those of human pancreatic intraepithelial neoplasia (PanIN) samples, demonstrating the relevance of this epigenetic transition state memory in human carcinogenesis. Finally, single-cell spatial transcriptomics revealed that these ADM transition cells were enriched for PI3K pathway and AP1 family members. CONCLUSIONS Our comprehensive study of DNA methylation in the acinar-ductal metaplasia transition state links epigenetic memory to cancer-related cell plasticity even in the absence of oncogenic mutation.
Collapse
Affiliation(s)
- Emily K W Lo
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD, 21205, USA
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument Street, Baltimore, MD, USA
| | - Adrian Idrizi
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument Street, Baltimore, MD, USA
| | - Rakel Tryggvadottir
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument Street, Baltimore, MD, USA
| | - Weiqiang Zhou
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Wenpin Hou
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Patrick Cahan
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD, 21205, USA.
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Andrew P Feinberg
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD, 21205, USA.
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, 1830 E. Monument Street, Baltimore, MD, USA.
| |
Collapse
|
14
|
Wu H, Shang J, Bao Y, Liu H, Zhang H, Xiao Y, Li Y, Huang Z, Cheng X, Ma Z, Zhang W, Mo P, Wang D, Zhang M, Zhan Y. Identification of a novel prognostic marker ADGRG6 in pancreatic adenocarcinoma: multi-omics analysis and experimental validation. Front Immunol 2025; 16:1530789. [PMID: 40226617 PMCID: PMC11986822 DOI: 10.3389/fimmu.2025.1530789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
Background Pancreatic adenocarcinoma (PAAD) ranks among the most lethal malignancies worldwide. Current treatment options have limited efficacy, underscoring the need for new therapeutic targets. Methods This study employed a multi-omics analytical framework to delve into the expression profiles and prognostic implications of ADGRG6 within the pan-cancer dataset of The Cancer Genome Atlas (TCGA) database, highlighting the prognostic value and potential carcinogenic role of ADGRG6 in PAAD, which was further validated using data from multiple PAAD cohorts in the Gene Expression Omnibus (GEO) database. To assess the role of ADGRG6 in the tumor microenvironment of PAAD, we evaluated immune infiltration using CIBERSORT, ssGSEA, xCell and Tracking Tumor Immunophenotype (TIP), and utilized single-cell sequencing data to explore cell-cell interactions. Further cellular and animal experiments, such as colony formation assay, transwell assay, western blot, real-time PCR, and tumor xenograft experiments, were used to investigate the effect of ADGRG6 on the proliferation, metastatic potential and immune marker expression of PAAD and the underlying mechanisms. Results ADGRG6 emerged as a potential prognostic biomarker and a therapeutic target for PAAD, which was further corroborated by data extracted from multiple PAAD cohorts archived in the GEO database. Single-cell sequencing and immune infiltration analyses predicted the positive correlation of ADGRG6 with the infiltration of immune cells and with the interaction between malignant cells and fibroblasts/macrophages within the PAAD microenvironment. In vitro cell assays demonstrated that ADGRG6 promoted the proliferation, metastatic potential and immune marker expression of PAAD cells by increasing protein level of mutated p53 (mutp53), which activated a spectrum of gain-of-functions to promote cancer progression via the EGFR, AMPK and NF-κB signaling cascades. Furthermore, subcutaneous xenograft experiments in mice demonstrated that ADGRG6 knockdown substantially suppressed the growth of engrafted PAAD tumors. Conclusions ADGRG6 may serve as a novel prognostic marker and a therapeutic targets for PAAD, playing a crucial role in the proliferation, metastasis, and immune marker regulation of PAAD through elevating protein level of mutated p53.
Collapse
Affiliation(s)
- Han Wu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Jin Shang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yuanyan Bao
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Huajie Liu
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Haoran Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yaosheng Xiao
- Department of Infectious Disease, Xiang’an Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
| | - Yangtaobo Li
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Zhaozhang Huang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xiaolei Cheng
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zixuan Ma
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wenqing Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Pingli Mo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Science, Xiamen University, Xiamen, Fujian, China
| | - Daxuan Wang
- Provincial College of Clinical Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Mingqing Zhang
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Yanyan Zhan
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
15
|
Guo X, Shao Y. Role of the oral-gut microbiota axis in pancreatic cancer: a new perspective on tumor pathophysiology, diagnosis, and treatment. Mol Med 2025; 31:103. [PMID: 40102723 PMCID: PMC11917121 DOI: 10.1186/s10020-025-01166-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: 10/06/2024] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
Pancreatic cancer, one of the most lethal malignancies, remains challenging due to late diagnosis, aggressive progression, and therapeutic resistance. Recent advances have revealed the presence of intratumoral microbiota, predominantly originating from the oral and gut microbiomes, which play pivotal roles in pancreatic cancer pathogenesis. The dynamic interplay between oral and gut microbial communities, termed the "oral-gut microbiota axis," contributes multifacetedly to pancreatic ductal adenocarcinoma (PDAC). Microbial translocation via anatomical or circulatory routes establishes tumor-resident microbiota, driving oncogenesis through metabolic reprogramming, immune regulation, inhibition of apoptosis, chronic inflammation, and dysregulation of the cell cycle. Additionally, intratumoral microbiota promote chemoresistance and immune evasion, further complicating treatment outcomes. Emerging evidence highlights microbial signatures in saliva and fecal samples as promising non-invasive diagnostic biomarkers, while microbial diversity correlates with prognosis. Therapeutic strategies targeting this axis-such as antibiotics, probiotics, and engineered bacteria-demonstrate potential to enhance treatment efficacy. By integrating mechanisms of microbial influence on tumor biology, drug resistance, and therapeutic applications, the oral-gut microbiota axis emerges as a critical regulator of PDAC, offering novel perspectives for early detection, prognostic assessment, and microbiome-based therapeutic interventions.
Collapse
Affiliation(s)
- Xuanchi Guo
- School of Stomatology, Shandong University, No. 44-1 Wenhua West Road, Jinan City, Shandong Province, China.
| | - Yuhan Shao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
16
|
Breaza GM, Closca RM, Cindrea AC, Hut FE, Cretu O, Sima LV, Rakitovan M, Zara F. Immunohistochemical Evaluation of the Tumor Immune Microenvironment in Pancreatic Ductal Adenocarcinoma. Diagnostics (Basel) 2025; 15:646. [PMID: 40075893 PMCID: PMC11899021 DOI: 10.3390/diagnostics15050646] [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: 02/07/2025] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma is an aggressive neoplasm with a complex carcinogenesis process that must be understood through the interactions between tumor cells and tumor microenvironment cells. Methods: This study was retrospective with a chronological extension period of 16 years and included 56 cases of pancreatic ductal adenocarcinoma. This study identified, quantified, and correlated the cells of the tumor immune microenvironment in pancreatic ductal adenocarcinoma with major prognostic factors as well as overall survival, using an extensive panel of immunohistochemical markers. Results: Three tumor immunotypes were identified: subtype A (hot immunotype), subtype B (intermediate immunotype), and subtype C (cold immunotype). Patients with immunotype C exhibit considerably higher rates of both pancreatic fistulas and acute pancreatitis. Immunotypes B and C significantly increased the risk of this complication by factors of 3.68 (p = 0.002) and 3.94 (p = 0.001), respectively. The estimated probabilities of fistula formation for each immunotype are as follows: 2.5% for immunotype A, 25% for immunotype B, and 28% for immunotype C. There was a statistically significant difference in median survival times according to tumor immunotype (p < 0.001). Specifically, patients with immunotype C tumors had a median survival time of only 120.5 days, compared to 553.5 days for those with immunotype A and 331.5 for immunotype B tumors. Conclusions: The identification of the immunotype of pancreatic ductal adenocarcinoma can be a predictive factor for the occurrence of complications such as pancreatic fistula as well as for overall survival.
Collapse
Affiliation(s)
- Gelu Mihai Breaza
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Raluca Maria Closca
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Department of Pathology, Emergency City Hospital, 300254 Timisoara, Romania
| | - Alexandru Cristian Cindrea
- Department of Surgery, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania;
- Emergency Department, Emergency Clinical Municipal Hospital, 300079 Timisoara, Romania
| | - Florin Emil Hut
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
- Center for Hepato-Bilio-Pancreatic Surgery, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania
| | - Octavian Cretu
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Laurentiu Vasile Sima
- University Clinic of Surgery I, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (F.E.H.); (O.C.); (L.V.S.)
| | - Marina Rakitovan
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Oro-Maxillo-Facial Surgery Clinic, Emergency City Hospital, 300062 Timisoara, Romania
| | - Flavia Zara
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania; (G.M.B.); (M.R.); (F.Z.)
- Department of Pathology, Emergency City Hospital, 300254 Timisoara, Romania
| |
Collapse
|
17
|
Dai D, Wang S, Li J, Zhao Y. Exploring radiation resistance-related genes in pancreatic cancer and their impact on patient prognosis and treatment. Front Immunol 2025; 16:1524798. [PMID: 40103813 PMCID: PMC11914796 DOI: 10.3389/fimmu.2025.1524798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Background Pancreatic cancer is a highly lethal disease with increasing incidence worldwide. Despite surgical resection being the main curative option, only a small percentage of patients are eligible for surgery. Radiotherapy, often combined with chemotherapy, remains a critical treatment, especially for locally advanced cases. However, pancreatic cancer's aggressiveness and partial radio resistance lead to frequent local recurrence. Understanding the mechanisms of radiotherapy resistance is crucial to improving patient outcomes. Methods Pancreatic cancer related gene microarray data were downloaded from GEO database to analyze differentially expressed genes before and after radiotherapy using GEO2R online tool. The obtained differentially expressed genes were enriched by GO and KEGG to reveal their biological functions. Key genes were screened by univariate and multivariate Cox regression analysis, and a risk scoring model was constructed, and patients were divided into high-risk group and low-risk group. Subsequently, Kaplan-Meier survival analysis was used to compare the survival differences between the two groups of patients, further analyze the differential genes of the two groups of patients, and evaluate their sensitivity to different drugs. Results Our model identified 10 genes associated with overall survival (OS) in pancreatic cancer. Based on risk scores, patients were categorized into high- and low-risk groups, with significantly different survival outcomes and immune profile characteristics. High-risk patients showed increased expression of pro-inflammatory immune markers and increased sensitivity to specific chemotherapy agents, while low-risk patients had higher expression of immune checkpoints (CD274 and CTLA4), indicating potential sensitivity to targeted immunotherapies. Cross-dataset validation yielded consistent AUC values above 0.77, confirming model stability and predictive accuracy. Conclusion This study provides a scoring model to predict radiotherapy resistance and prognosis in pancreatic cancer, with potential clinical application for patient stratification. The identified immune profiles and drug sensitivity variations between risk groups highlight opportunities for personalized treatment strategies, contributing to improved management and survival outcomes in pancreatic cancer.
Collapse
Affiliation(s)
- Dong Dai
- Department of Nuclear Medicine, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for China, Tianjin, China
| | - Sen Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jiaze Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yu Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
18
|
Ding J, Xie Y, Liu Z, Zhang Z, Ni B, Yan J, Zhou T, Hao J. Hypoxic and Acidic Tumor Microenvironment-Driven AVL9 Promotes Chemoresistance of Pancreatic Ductal Adenocarcinoma via the AVL9-IκBα-SKP1 Complex. Gastroenterology 2025; 168:539-555.e5. [PMID: 39566663 DOI: 10.1053/j.gastro.2024.10.042] [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: 01/03/2024] [Revised: 09/24/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND & AIMS Gemcitabine combined with albumin-paclitaxel (AG) is a crucial therapeutic option for pancreatic ductal adenocarcinoma (PDAC). However, the response to chemotherapy is relatively poor, with rapid development of resistance. The aim of this study was to explore the mechanism of resistance to AG and to develop strategies that can sensitize the AG regimen. METHODS We used organoid models, patient-derived xenografts, and genetically engineered mouse models in our study. Chromatin immunoprecipitation, double luciferase assay, co-immunoprecipitation, and far-western blotting analysis were performed to investigate the mechanism. The AVL9 inhibitors were identified through protein structure analysis and molecular docking analysis, and their efficacy was verified in patient-derived xenografts, patient-derived organoids-based xenograft, and KPC models. RESULTS Through multistrategy screening, we identified AVL9 as a key target for AG resistance in PDAC. Its tumor-promoting effects were confirmed in our clinical cohorts. Mechanistically, HIF-1α, a hypoxia-related transcription factor, drives the expression of AVL9. AVL9 acts as a scaffold that facilitates the binding of IκBα to SKP1, leading to enhanced ubiquitination and degradation of IκBα, which further activates the nuclear factor-κB pathway. The potential AVL9-targeting inhibitor, Edotecarin, was shown to reverse AG chemo-resistance in PDAC. CONCLUSION AVL9 expression is driven by HIF-1α in PDAC. The physical interaction of AVL9, IκBα, and SKP1 provides a novel molecular mechanism for the abnormal activation of the nuclear factor-κB pathway. Therefore, the AVL9-targeting drug Edotecarin could be a promising therapeutic strategy for sensitizing PDAC to AG.
Collapse
Affiliation(s)
- Jinsheng Ding
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China; Department of Breast Oncoplastic Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, People's Republic of China
| | - Yongjie Xie
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Ziyun Liu
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Zhaoyu Zhang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Bo Ni
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Jingrui Yan
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China
| | - Tianxing Zhou
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China.
| | - Jihui Hao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, People's Republic of China.
| |
Collapse
|
19
|
Lakis V, Chan NL, Lyons R, Blackburn N, Nguyen TH, Chang C, Masel A, West NP, Boyle GM, Patch AM, Gill AJ, Nones K. Spatial Transcriptomics Reveals Novel Mechanisms Involved in Perineural Invasion in Pancreatic Ductal Adenocarcinomas. Cancers (Basel) 2025; 17:852. [PMID: 40075699 PMCID: PMC11899704 DOI: 10.3390/cancers17050852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a high incidence of perineural invasion (PNI), a pathological feature of the cancer invasion of nerves. PNI is associated with a poor prognosis, local recurrence and cancer pain. It has been suggested that interactions between nerves and the tumor microenvironment (TME) play a role in PDAC tumorigenesis. METHODS Here, we used Nanostring GeoMx Digital Spatial Profiler to analyze the whole transcriptome of both cancer and nerve cells in the microenvironment of PNI and non-PNI foci from 13 PDAC patients. CONCLUSIONS We identified previously reported pathways involved in PNI, including Axonal Guidance and ROBO-SLIT Signaling. Spatial transcriptomics highlighted the role of PNI foci in influencing the immune landscape of the TME and similarities between PNI and nerve injury response. This study revealed that endocannabinoid and polyamine metabolism may contribute to PNI, cancer growth and cancer pain. Key members of these pathways can be targeted, offering potential novel research avenues for exploring new cancer treatment and/or pain management options in PDAC.
Collapse
Affiliation(s)
- Vanessa Lakis
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia; (V.L.); (T.H.N.); (C.C.); (A.M.); (G.M.B.); (A.-M.P.)
| | - Noni L Chan
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, NSW 2065, Australia; (N.L.C.); (A.J.G.)
| | - Ruth Lyons
- Australian Pancreatic Cancer Genome Initiative (APGI), Kinghorn Cancer Centre, Sydney, NSW 2010, Australia; (R.L.); (N.B.)
| | - Nicola Blackburn
- Australian Pancreatic Cancer Genome Initiative (APGI), Kinghorn Cancer Centre, Sydney, NSW 2010, Australia; (R.L.); (N.B.)
| | - Tam Hong Nguyen
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia; (V.L.); (T.H.N.); (C.C.); (A.M.); (G.M.B.); (A.-M.P.)
| | - Crystal Chang
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia; (V.L.); (T.H.N.); (C.C.); (A.M.); (G.M.B.); (A.-M.P.)
| | - Andrew Masel
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia; (V.L.); (T.H.N.); (C.C.); (A.M.); (G.M.B.); (A.-M.P.)
| | - Nicholas P. West
- Griffith Health, Griffith University, Gold Coast, QLD 4215, Australia;
| | - Glen M. Boyle
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia; (V.L.); (T.H.N.); (C.C.); (A.M.); (G.M.B.); (A.-M.P.)
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Kelvin Grove, QLD 4000, Australia
| | - Ann-Marie Patch
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia; (V.L.); (T.H.N.); (C.C.); (A.M.); (G.M.B.); (A.-M.P.)
| | - Anthony J. Gill
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, NSW 2065, Australia; (N.L.C.); (A.J.G.)
- Australian Pancreatic Cancer Genome Initiative (APGI), Kinghorn Cancer Centre, Sydney, NSW 2010, Australia; (R.L.); (N.B.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW 2065, Australia
| | - Katia Nones
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia; (V.L.); (T.H.N.); (C.C.); (A.M.); (G.M.B.); (A.-M.P.)
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
- Faculty of Health, Medicine and Behavioural Sciences/PA Southside Clinical Unit, The University of Queensland, Brisbane, QLD 4102, Australia
| |
Collapse
|
20
|
Fasala A, Carr M, Surjan Y, Daghigh P, de Leon J, Burns A, Batumalai V. Intrafraction motion and impact of margin reduction for MR-Linac online adaptive radiotherapy for pancreatic cancer treatments. J Med Radiat Sci 2025; 72:17-24. [PMID: 39397350 PMCID: PMC11909694 DOI: 10.1002/jmrs.832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/28/2024] [Indexed: 10/15/2024] Open
Abstract
INTRODUCTION Online adaptive radiotherapy is well suited for stereotactic ablative radiotherapy (SABR) in pancreatic cancer due to considerable intrafractional tumour motion. This study aimed to assess intrafraction motion and generate adjusted planning target volume (PTV) margins required for online adaptive radiotherapy in pancreatic cancer treatment using abdominal compression on the magnetic resonance linear accelerator (MR-Linac). METHODS Motion monitoring images obtained from 67 fractions for 15 previously treated pancreatic cancer patients were analysed. All patients received SABR (50 Gy in five fractions) on the MR-Linac using abdominal compression. The analysis included quantification of intrafraction motion, leading to the development of adjusted PTV margins. The dosimetric impact of implementing the adjusted PTV was then evaluated in a cohort of 20 patients. RESULTS Intrafraction motion indicated an average target displacement of 1-3 mm, resulting in an adjusted PTV margin of 2 mm in the right-left and superior-inferior directions, and 3 mm in the anterior-posterior direction. Plans incorporating these adjusted margins consistently demonstrated improved dose to target volumes, with improvements averaging 1.5 Gy in CTV D99%, 4.9 Gy in PTV D99% and 1.2 Gy in PTV-high D90%, and better sparing of the organs at risk (OAR). CONCLUSIONS The improved target volume coverage and reduced OAR dose suggest potential for reducing current clinical margins for MR-Linac treatment. However, it is important to note that decreasing margins may reduce safeguards against geographical misses. Nonetheless, the continued integration of gating systems on MR-Linacs could provide confidence in adopting reduced margins.
Collapse
Affiliation(s)
- Ashleigh Fasala
- GenesisCare, Sydney, New South Wales, Australia
- College of Health, Medicine and Wellbeing, School of Health Sciences, Global Centre for Research and Training in Radiation Oncology, The University of Newcastle, Callaghan, New South Wales, Australia
| | | | - Yolanda Surjan
- College of Health, Medicine and Wellbeing, School of Health Sciences, Global Centre for Research and Training in Radiation Oncology, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Parmoun Daghigh
- School of Physics, University of Sydney, Sydney, New South Wales, Australia
| | | | - Abbey Burns
- GenesisCare, Sydney, New South Wales, Australia
| | - Vikneswary Batumalai
- GenesisCare, Sydney, New South Wales, Australia
- The George Institute for Global Health, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
21
|
Daya T, Breytenbach A, Gu L, Kaur M. Cholesterol metabolism in pancreatic cancer and associated therapeutic strategies. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159578. [PMID: 39542394 DOI: 10.1016/j.bbalip.2024.159578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Pancreatic cancer remains one of the most lethal cancers due to late diagnosis and high chemoresistance. Despite recent progression in the development of chemotherapies, immunotherapies, and potential nanoparticles-based approaches, the success rate of therapeutic response is limited which is further compounded by cancer drug resistance. Understanding of emerging biological and molecular pathways causative of pancreatic cancer's aggressive and chemoresistance is vital to improve the effectiveness of existing therapeutics and to develop new therapies. One such under-investigated and relatively less explored area of research is documenting the effect that lipids, specifically cholesterol, and its metabolism, impose on pancreatic cancer. Dysregulated cholesterol metabolism has a profound role in supporting cellular proliferation, survival, and promoting chemoresistance and this has been well established in various other cancers. Thus, we aimed to provide an in-depth review focusing on the significance of cholesterol metabolism in pancreatic cancer and relevant genes at play, molecular processes contributing to cellular cholesterol homeostasis, and current research efforts to develop new cholesterol-targeting therapeutics. We highlight the caveats, weigh in different experimental therapeutic strategies, and provide possible suggestions for future research highlighting cholesterol's importance as a therapeutic target against pancreatic cancer resistance and cancer progression.
Collapse
Affiliation(s)
- Tasvi Daya
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Andrea Breytenbach
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Liang Gu
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Mandeep Kaur
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa.
| |
Collapse
|
22
|
Lasota M, Jankowski D, Wiśniewska A, Szeleszczuk Ł, Misterka-Kozaka A, Kaczor-Kamińska M, Zarzycka M, Patena M, Brzozowski T. Interaction of Avapritinib with Congo Red in Pancreatic Cancer Cells: Molecular Modeling and Biophysical Studies. Int J Mol Sci 2025; 26:1980. [PMID: 40076604 PMCID: PMC11901030 DOI: 10.3390/ijms26051980] [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: 01/15/2025] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Pancreatic cancer is a malignant tumor with one of the worst prognoses among solid tumors, characterized by resistance to treatment. Therefore, there is an urgent need for new methods of targeted therapy. Previous studies have shown that the overexpression of receptor tyrosine kinases such as c-KIT or PDGFR can increase proliferation, migration, and invasion of cancer cells. The aim of our study was to analyze aggregates between a supramolecular carrier (Congo red, CR) and a tyrosine kinase inhibitor (BLU-258) as well as to investigate the effect of the free inhibitor and its aggregate with Congo red (CR-BLU-258) on selected properties of pancreatic cells, including these cells' viability and three-dimensional cell spheroid cultures. To better understand the interactions between Congo red and BLU-258, we used molecular modeling in addition to biophysical methods. These attempts allowed us to determine the optimal molar ratio, which we used for in vitro studies on pancreatic cancer cell lines. A significantly greater decrease in the viability of the tested 3D cultures was observed after 48 h of incubation with CR-BLU-258, which resulted in a lower IC50 value for the tested co-aggregate compared with BLU-258 alone. Moreover, a higher resistance of PANC-1 and BxPC3 spheroid cells to the tested compounds was noted compared with the 2D culture model. A significantly lower response was observed in 3D cell cultures (BxPC3 and PANC-1) treated with BLU-258 alone compared with the 2D culture. Thus, our results showed that both BLU-258 (alone) and in its co-aggregate with Congo red exhibit anticancer activity, inhibiting the growth of pancreatic cancer cells and reducing their viability, survival, and migration. Both tested compounds also affected the phosphorylation of the selected signaling proteins. We conclude that the selected tyrosine kinase inhibitor (alone) and in its co-aggregate with Congo red exhibit anticancer activity and should be considered as a novel effective therapy against pancreatic cancer.
Collapse
Affiliation(s)
- Małgorzata Lasota
- Center for Biomedicine and Interdisciplinary Sciences, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Daniel Jankowski
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Anna Wiśniewska
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
| | - Łukasz Szeleszczuk
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097 Warsaw, Poland;
| | - Anna Misterka-Kozaka
- Center for Biomedicine and Interdisciplinary Sciences, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Marta Kaczor-Kamińska
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, 31-034 Krakow, Poland; (M.K.-K.); (M.Z.)
| | - Marta Zarzycka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, 31-034 Krakow, Poland; (M.K.-K.); (M.Z.)
| | - Maksym Patena
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Tomasz Brzozowski
- Center for Biomedicine and Interdisciplinary Sciences, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland
| |
Collapse
|
23
|
Gupta T, Murtaza M. Advancing Targeted Therapies in Pancreatic Cancer: Leveraging Molecular Aberrations for Therapeutic Success. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025:S0079-6107(25)00016-1. [PMID: 39988056 DOI: 10.1016/j.pbiomolbio.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 02/03/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Pancreatic cancer is one of the most deadly with poor prognosis and overall survival rate due to the dense stroma in the tumors which often is challenging for the delivery of drug to penetrate deep inside the tumor bed and usually results in the progression of cancer. The conventional treatment such as chemotherapy, radiotherapy or surgery shows a minimal benefit in the survival due to the drug resistance, poor penetration, less radiosensitivity or recurrence of tumor. There is an urgent demand to develop molecular- level targeted therapies to achieve therapeutic efficacy in the pancreatic ductal adenocarcinoma (PDAC) patients. The precision oncology focuses on the unique attributes of the patient such as epigenome, proteome, genome, microbiome, lifestyle and diet habits which contributes to promote oncogenesis. The targeted therapy helps to target the mutated proteins responsible for controlling growth, division and metastasis of tumor in the cancer cells. It is very important to consider all the attributes of the patient to provide the suitable personalized treatment to avoid any severe side effects. In this review, we have laid emphasis on the precision medicine; the utmost priority is to improve the survival of cancer patients by targeting molecular mutations through transmembrane proteins, inhibitors, signaling pathways, immunotherapy, gene therapy or the use of nanocarriers for the delivery at the tumor site. It will become beneficial therapeutic window to be considered for the advanced stage pancreatic cancer patients to prolong their survival rate.
Collapse
Affiliation(s)
- Tanvi Gupta
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan.
| | - Mohd Murtaza
- Fermentation & Microbial Biotechnology Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180016, India.
| |
Collapse
|
24
|
Sherpally D, Manne A. Advancing Immunotherapy in Pancreatic Cancer: A Brief Review of Emerging Adoptive Cell Therapies. Cancers (Basel) 2025; 17:589. [PMID: 40002184 PMCID: PMC11853216 DOI: 10.3390/cancers17040589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/28/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Pancreatic cancer has the lowest 5-year survival rate (13%) among major cancers and is the third leading cause of cancer-related deaths in the United States. The high lethality of this cancer is attributed to its insidious onset, late-stage diagnosis, rapid progression, and limited treatment options. Addressing these challenges requires a deeper understanding of the complex tumor microenvironment to identify novel therapeutic targets. Newer approaches like adoptive cell therapy have shown remarkable success in treating hematological malignancies, but their application in solid tumors, particularly pancreatic cancer, is still in the early stages of development. ACT broadly involves isolating immune cells (T lymphocytes, Natural Killer cells, and macrophages) from the patient, followed by genetic engineering to enhance and mount a specific anti-tumor response. Various ACT modalities are under investigation for pancreatic cancer, including chimeric antigen receptor T cells (CAR-T), chimeric antigen receptor NK cells (CAR-NK), tumor-infiltrating lymphocytes (TIL), T-cell receptor (TCR)-engineered T cells, and cytokine-induced killer cells (CIK). Major hurdles have been identifying actionable tumor antigens and delivering focused cellular therapies to overcome the immunosuppressive and dense fibrotic stroma surrounding the pancreatic cancer. Further studies are needed to explore the limitations faced by cellular therapy in pancreatic cancer and identify novel combination treatment approaches in order to improve clinical outcomes.
Collapse
Affiliation(s)
- Deepak Sherpally
- Department of Internal Medicine, New York Medical College, Metropolitan, New York, NY 10029, USA
| | - Ashish Manne
- Department of Internal Medicine, Division of Medical Oncology, The Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA;
| |
Collapse
|
25
|
Mohamadkhani A, Ghanbari R, Shakeri R, Mohammadkhani MA, Pourshams A. Causal Exposures in Pancreatic Cancer Incidence: Insights From Mendelian Randomization Studies. JGH Open 2025; 9:e70105. [PMID: 39906083 PMCID: PMC11790352 DOI: 10.1002/jgh3.70105] [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: 11/02/2024] [Revised: 01/04/2025] [Accepted: 01/20/2025] [Indexed: 02/06/2025]
Abstract
Aim Pancreatic cancer, marked by its high lethality and poor 5-year survival rate, requires a thorough understanding of its risk factors and etiological mechanisms. In this review, we collected the latest findings from Mendelian randomization (MR) studies to identify potential causal factors for pancreatic cancer. Method and Results The present analysis encompasses MR studies on the gut and oral microbiomes, non-malignant phenotypes, blood metabolites, immune cells, and chronic inflammation. Specific gut and oral microbiome species have been identified as potential causal factors for pancreatic cancer, some with protective effects, and others increasing the risk. The review also highlights causal associations between obesity, type 2 diabetes, and pancreatic cancer, as well as the impact of blood metabolites and immune cell phenotypes on disease risk. Additionally, it investigates the causal effects of inflammatory bowel disease, showing a significant risk increase associated with Crohn's disease. Conclusion These insights emphasize the need for interdisciplinary research and personalized medicine to enhance prevention and treatment strategies for pancreatic cancer.
Collapse
Affiliation(s)
- Ashraf Mohamadkhani
- Liver and Pancreatobiliary Diseases Research Center; Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical SciencesTehranIran
| | - Reza Ghanbari
- Gene Therapy Research Center, Digestive Diseases Research Institute, Tehran University of Medical SciencesTehranIran
| | - Ramin Shakeri
- Digestive Oncology Research Center, Digestive Diseases Research Institute, Tehran University of Medical SciencesTehranIran
| | | | - Akram Pourshams
- Liver and Pancreatobiliary Diseases Research Center; Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical SciencesTehranIran
- Digestive Oncology Research Center, Digestive Diseases Research Institute, Tehran University of Medical SciencesTehranIran
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
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
|
29
|
Maebashi M, Miyake K, Yamamoto J, Sahara K, Akiyama T, Kimura Y, Endo I. Methionine restriction inhibits pancreatic cancer proliferation while suppressing JAK2/STAT3 pathway. Pancreatology 2025; 25:108-117. [PMID: 39668011 DOI: 10.1016/j.pan.2024.11.023] [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: 03/29/2024] [Revised: 10/30/2024] [Accepted: 11/28/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Methionine restriction (MR) has been demonstrated to exhibit anti-tumor effects in various types of cancer, including pancreatic cancer (PC). However, the detailed mechanism induced by MR remains still unclear. This study aims to reveal the underlying mechanism of MR on PC by proteomic analysis. MATERIAL & METHODS Human PC cell lines were cultured in both standard and MR media to evaluate the effect of MR. The differences in protein expression were evaluated through proteomic analysis. Ingenuity Pathway Analysis (IPA) was performed to identify proteins potentially associated with tumor growth in vitro. The proteins associated with the anti-tumor effect were validated using western blotting, real-time PCR, and ELISA. An experimental model involving subcutaneous PC mice was established for the assessment of the effectiveness of the MR diet and the expression of target proteins through immunohistochemical staining. RESULTS Cell proliferation was suppressed in the MR media compared to the standard media. IPA analysis showed that STAT3 was decreased in the Apoptotic Pathway of Pancreatic Cancer Cell lines in the MR group. Western blotting showed MR decreased STAT3 expression. Real-time PCR showed that MR decreased JAK2 and STAT3 mRNA expression in Panc-1 and Mia-PaCa 2, but not in Capan-1. ELISA revealed that NF-kB expression was decreased in the MR group. In the in vivo study, the final estimated tumor volume in the MR group was significantly lower than the control group (p < 0.01). Immunostaining of resected specimens showed that STAT3 expression was suppressed in the MR group. CONCLUSION MR suppressed the JAK2/STAT3 pathway and decreased NF-kB in some PC cell lines.
Collapse
Affiliation(s)
- Manabu Maebashi
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kentaro Miyake
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Jun Yamamoto
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kota Sahara
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomoko Akiyama
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Yayoi Kimura
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
30
|
Bush N, Yazici C, Han SY, Lee PJ, Abu-El-Haija M, Akshintala VS. Assessment of Medical Pancreatology Fellowship Training in the United States: A Collaborative Alliance for Pancreatic Education and Research (CAPER) Initiative. Pancreas 2025; 54:e166-e167. [PMID: 39928894 DOI: 10.1097/mpa.0000000000002406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Affiliation(s)
- Nikhil Bush
- From the Department of Medicine, Division of Gastroenterology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Cemal Yazici
- Department of Medicine, Division of Gastroenterology, University of Illinois Chicago, Chicago, IL
| | - Samuel Y Han
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Peter J Lee
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Maisam Abu-El-Haija
- Department of Pediatrics, Division of Pediatric Gastroenterology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Venkata S Akshintala
- From the Department of Medicine, Division of Gastroenterology, Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
31
|
Ferreira N, Kulkarni A, Agorku D, Midelashvili T, Hardt O, Legler TJ, Ströbel P, Conradi LC, Alves F, Ramos-Gomes F, Markus MA. OrganoIDNet: a deep learning tool for identification of therapeutic effects in PDAC organoid-PBMC co-cultures from time-resolved imaging data. Cell Oncol (Dordr) 2025; 48:101-122. [PMID: 38805131 PMCID: PMC11850476 DOI: 10.1007/s13402-024-00958-2] [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] [Accepted: 05/06/2024] [Indexed: 05/29/2024] Open
Abstract
PURPOSE Pancreatic Ductal Adenocarcinoma (PDAC) remains a challenging disease due to its complex biology and aggressive behavior with an urgent need for efficient therapeutic strategies. To assess therapy response, pre-clinical PDAC organoid-based models in combination with accurate real-time monitoring are required. METHODS We established stable live-imaging organoid/peripheral blood mononuclear cells (PBMCs) co-cultures and introduced OrganoIDNet, a deep-learning-based algorithm, capable of analyzing bright-field images of murine and human patient-derived PDAC organoids acquired with live-cell imaging. We investigated the response to the chemotherapy gemcitabine in PDAC organoids and the PD-L1 inhibitor Atezolizumab, cultured with or without HLA-matched PBMCs over time. Results obtained with OrganoIDNet were validated with the endpoint proliferation assay CellTiter-Glo. RESULTS Live cell imaging in combination with OrganoIDNet accurately detected size-specific drug responses of organoids to gemcitabine over time, showing that large organoids were more prone to cytotoxic effects. This approach also allowed distinguishing between healthy and unhealthy status and measuring eccentricity as organoids' reaction to therapy. Furthermore, imaging of a new organoids/PBMCs sandwich-based co-culture enabled longitudinal analysis of organoid responses to Atezolizumab, showing an increased potency of PBMCs tumor-killing in an organoid-individual manner when Atezolizumab was added. CONCLUSION Optimized PDAC organoid imaging analyzed by OrganoIDNet represents a platform capable of accurately detecting organoid responses to standard PDAC chemotherapy over time. Moreover, organoid/immune cell co-cultures allow monitoring of organoid responses to immunotherapy, offering dynamic insights into treatment behavior within a co-culture setting with PBMCs. This setup holds promise for real-time assessment of immunotherapeutic effects in individual patient-derived PDAC organoids.
Collapse
Affiliation(s)
- Nathalia Ferreira
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Ajinkya Kulkarni
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - David Agorku
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Teona Midelashvili
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany
| | - Olaf Hardt
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Tobias J Legler
- Department of Transfusion Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Lena-Christin Conradi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany
| | - Frauke Alves
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
- Clinic of Hematology and Medical Oncology, Department of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Fernanda Ramos-Gomes
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - M Andrea Markus
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
32
|
Kudaravalli P, Singh S, Vinayek R, Aswath G, Crinò SF, Machicado J, Facciorusso A. Reducing the incidence of pancreatic cancer through radiofrequency ablation of mucinous cystic neoplasms: What is the evidence? Best Pract Res Clin Gastroenterol 2025; 74:101986. [PMID: 40210331 DOI: 10.1016/j.bpg.2025.101986] [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: 10/19/2024] [Revised: 01/08/2025] [Accepted: 01/24/2025] [Indexed: 03/04/2025]
Abstract
Pancreatic cystic lesions are mostly discovered incidentally during cross-sectional imaging for other indications. They range from benign cysts to premalignant lesions, posing a risk for development of pancreatic cancer. Surveillance of cysts with low-risk features and surgical resection of high-risk ones has been the standard of care. However, the lack of consensus between national society guidelines results in discordant management and follow-up. One of the upcoming therapeutic modalities for pancreatic cysts, including mucinous cystic neoplasms (MCN), is endoscopic ultrasound (EUS)-guided interventions, including injection of chemotherapeutic agents or EUS-guided radiofrequency ablation (EUS-RFA). EUS-RFA is emerging as a promising minimally invasive tool with the need for further investigation to understand its clinical utility. This review discusses the physiology, clinical studies, adverse events, and future perspectives of RFA for various pancreatic lesions, with particular regard with MCN.
Collapse
Affiliation(s)
- Pujitha Kudaravalli
- Gastroenterology & Hepatology, Lahey Hospital & Medical Center, Burlington, MA, United States.
| | - Sahib Singh
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, 21215, United States.
| | - Rakesh Vinayek
- Gastroenterology & Hepatology, Sinai Hospital of Baltimore, Baltimore, MD, 21215, United States.
| | - Ganesh Aswath
- Gastroenterology & Hepatology, State University of New York Upstate Medical University, Syracuse, NY, United States.
| | - Stefano Francesco Crinò
- Diagnostic and Interventional Endoscopy of Pancreas, The Pancreas Institute, University of Verona, 37134, Verona, Italy.
| | - Jorge Machicado
- Division of Gastroenterology, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States.
| | - Antonio Facciorusso
- Gastroenterology Unit, Department of Experimental Medicine, Università del Salento, 73100, Lecce, Italy.
| |
Collapse
|
33
|
Sahu SK, Prabhakar PK, Vyas M. Therapeutical potential of natural products in treatment of pancreatic cancer: a review. Mol Biol Rep 2025; 52:179. [PMID: 39888508 DOI: 10.1007/s11033-025-10287-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/22/2025] [Indexed: 02/01/2025]
Abstract
Pancreatic cancer remains as global health challenge, ranking as the seventh leading cause of cancer-related deaths worldwide with high mortality rates and a low five-year survival rate. Despite advancements in conventional therapies, including surgery, chemotherapy, and radiation, the overall survival rates for pancreatic cancer patients have shown minimal improvement. Consequently, there is an urgent need for alternative therapeutic strategies. The search for effective treatments has increasingly turned towards natural products, which offer a diverse array of bioactive compounds with potential anticancer properties. All the natural products, derived from plants, marine organisms, and microorganisms, have emerged as promising candidates in cancer treatment. The review explores the potential role of various natural compounds such as polyphenols, alkaloids, terpenoids, and flavonoids in pancreatic cancer management. With over 60% of cancer medications in clinical trials having natural origins, the review underscores the importance of exploring these compounds for their chemopreventive potential. It covers the epidemiological, molecular pathways influenced by these natural products (such as apoptosis, cell cycle regulation and signaling pathways) and therapeutic aspects aims to contribute to the ongoing efforts in understanding and addressing the complexities of pancreatic cancer. Overall, this review highlights the urgency of developing novel therapeutic strategies and incorporating natural compounds into current treatment modalities to improve outcomes for pancreatic cancer patients.
Collapse
Affiliation(s)
- Sanjeev Kumar Sahu
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| | - Pranav Kumar Prabhakar
- Research and Development Cell, Parul University, P.O. Limda, Dist. Vadodara, Ta.Waghodia, Gujarat, 391760, India
| | - Manish Vyas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| |
Collapse
|
34
|
Baghel K, Mehrotra S, Prajapati VK. Revolutionizing pancreatic cancer treatment with CAR-T therapy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:331-353. [PMID: 39978971 DOI: 10.1016/bs.apcsb.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Pancreatic cancer remains one of the most lethal malignancies, with a five-year survival rate among the lowest of all cancers. This poor prognosis is largely due to the aggressive nature of the disease and its resistance to conventional treatments such as surgery, chemotherapy, and radiation therapy. Chimeric antigen receptor (CAR) T-cell therapy, a novel immunotherapeutic approach leverages the patient's own immune system to specifically target and eliminate cancer cells by genetically engineering T cells to express CARs that recognize tumor-specific antigens. While CAR-T therapy has demonstrated remarkable success in treating hematologic malignancies, its application to solid tumors like pancreatic cancer presents significant challenges. Recent advancements in CAR-T cell design, like the addition of co-stimulatory domains and dual-targeting CARs, have enhanced their efficacy against solid tumors. Additionally, strategies to modify the tumor microenvironment (TME), such as combining CAR-T therapy with immune checkpoint inhibitors and cytokine modulation, are being investigated to boost CAR-T cell activity against pancreatic cancer. Early-phase clinical trials targeting antigens such as carcinoembryonic antigen (CEA) and mesothelin (MSLN) in pancreatic cancer have yielded encouraging results, though obstacles like antigen escape and limited T-cell persistence remain significant challenges. This chapter outlines the current state of CAR-T therapy for pancreatic cancer, focusing on the emerging approaches to address these obstacles and underscore the potential of CAR-T therapy to transform the future of pancreatic cancer treatment.
Collapse
Affiliation(s)
- Kirti Baghel
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
35
|
Yu W, Zhou D, Meng F, Wang J, Wang B, Qiang J, Shen L, Wang M, Fang H. The global, regional burden of pancreatic cancer and its attributable risk factors from 1990 to 2021. BMC Cancer 2025; 25:186. [PMID: 39891086 PMCID: PMC11786447 DOI: 10.1186/s12885-025-13471-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 01/07/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Pancreatic cancer is the 12th most common type of cancer, and the sixth leading cause of cancer-related mortality, worldwide. Up-to-date statistics on pancreatic cancer would provide us with a better understanding of epidemiology and identify the causative risk factors for the prevention of this disease. METHODS The degree and change patterns of exposure as well as the attributable cancer burden, including incidence, mortality, disability-adjusted life years (DALYs), and prevalence in global and regional, by sex, age, year, for pancreatic cancer, with the data extracted from the Global Burden of Diseases Study (GBD) 2021. All data analyses were conducted using linear regression analysis and the Joinpoint software (version 5.0.1). RESULTS In 2021, 508,533 new cases of pancreatic cancer have been reported; the mortality and prevalence rate increased to 5.95, and 5.12 respectively; and the global DALYs rate increased to 130.33 this year. Besides, the pancreatic cancer-associated rates of incidence, mortality, DALYs, and prevalence were higher in males than in females. In addition, these indicators in the high SDI (Sociodemographic index) region were higher than the global mean. To date, the high fasting plasma glucose remained the major risk factor that influenced the incidence, mortality, DALYs, and prevalence of pancreatic cancer, followed by tobacco and high body mass index (BMI). CONCLUSIONS Results of this study suggest that the burden of pancreatic cancer is increasing generally, therefore, more attention and measures should be taken to cope with this situation.
Collapse
Affiliation(s)
- Weidong Yu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Danyi Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Fanhao Meng
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jinjing Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Bo Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jianling Qiang
- Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, 322100, China
| | - Lijun Shen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Maofeng Wang
- Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, 322100, China.
| | - Hezhi Fang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China.
| |
Collapse
|
36
|
Helal NS, Maher S, Samir S, Elmeligy HA, Aboul-Ezz MA, Aboushousha T, Moussa M. Assessing the diagnostic potential of SATB2 and β-catenin as biomarkers and therapeutic targets in pancreatic ductal adenocarcinoma. J Cancer Res Clin Oncol 2025; 151:56. [PMID: 39878802 PMCID: PMC11779791 DOI: 10.1007/s00432-024-06055-z] [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: 09/05/2024] [Accepted: 12/03/2024] [Indexed: 01/31/2025]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is associated with poor prognosis. The roles of the transcription factor special AT-rich binding protein-2 (SATB2) and β-catenin in PDAC have been a subject of controversy. We aimed to assess the diagnostic and prognostic impact of SATB2 and β-catenin in PDAC. METHODS We analyzed 44 paraffin-embedded tissue blocks along with corresponding blood and pancreatic tissues. We evaluated SATB2 expression using immunohistochemistry (IHC) and enzyme-linked immunosorbent assay (ELISA). β-catenin was assessed using IHC and real-time polymerase chain reaction (qPCR). RESULTS High SATB2 expression and low β-catenin expression were associated with a poor prognosis in PDAC, including advanced pathological tumor stage (pT-stage), pathological lymph node stage (pN-stage), and TNM stage. We found a positive correlation between SATB2 expression assessed by IHC and the concentration of SATB2 in both serum and tissue samples measured by ELISA. We observed a positive correlation between β-catenin expression assessed by IHC and β-catenin levels measured by qPCR. CONCLUSIONS SATB2 and β-catenin could provide valuable insights into the development of pancreatic cancer, and targeting them may be beneficial for the prevention and treatment of PDAC. The levels of SATB2 in serum show promise for the diagnosis and tumor invasion of pancreatic cancer.
Collapse
Affiliation(s)
- Noha Said Helal
- Department of Pathology, Theodor Bilharz Research Institute, Giza, 12411, Egypt
| | - Sara Maher
- Department of Immunology, Theodor Bilharz Research Institute, Giza, 12411, Egypt
| | - Safia Samir
- Department of Biochemistry and Molecular Biology, Theodor Bilharz Research Institute, Giza, Egypt.
| | - Hesham A Elmeligy
- Department of Surgery, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mohammed A Aboul-Ezz
- Department of Hepatology and Gastroenterology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Tarek Aboushousha
- Department of Pathology, Theodor Bilharz Research Institute, Giza, 12411, Egypt
| | - Mona Moussa
- Department of Pathology, Theodor Bilharz Research Institute, Giza, 12411, Egypt
| |
Collapse
|
37
|
Pekmezci Y, Ergun S, Turgut BC, Dumur S, Sayili U, Uzun H, Pekmezci S, Velidedeoglu M. The Role of Resolvin D1 in the Differential Diagnosis of Pancreatic Ductal Adenocarcinoma and Acute Pancreatitis: A Case-Control Study. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:168. [PMID: 40005286 PMCID: PMC11857486 DOI: 10.3390/medicina61020168] [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: 12/03/2024] [Revised: 01/09/2025] [Accepted: 01/18/2025] [Indexed: 02/27/2025]
Abstract
Background and Objectives: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy characterized by a dense desmoplastic stroma with a poor prognosis. The aim of this study was to investigate whether resolvin (Rv) D1 could be used as a potential serum biomarker to discriminate between PDAC and acute pancreatitis (AP). Materials and Methods: In total, 67 patients were enrolled in the present study, including 21 patients with resectable PDAC, 23 patients with metastatic PDAC, 23 patients with AP, and a control group of 21 healthy individuals. RvD1 levels of PDAC patients were also analyzed through ELISA at the 6th postoperative month. Results: The mean RvD1 was 1169.24 ± 285.99 in the control group, 885.04 ± 134.25 in the AP group, 728.57 ± 140.1 in the PDAC group, and 670.09 ± 105.6 in the metastatic pancreatic cancer (PC) group. RvD1 was significantly lower in PDAC and metastatic PC groups compared to controls and patients with AP, while it was significantly lower in patients with AP compared to the control groups. Postoperative RvD1 levels of patients with PDAC were significantly higher than preoperative levels (728.57 ± 140.1 vs. 885.43 ± 275.57). In the ROC analysis, when the cut-off value for serum RvD1 level was 825 ng/L, it was found to predict PDAC from metastatic PC with 84.1% sensitivity and 81.8% specificity. Conclusions: Serum RvD1 is a new biomarker for the detection of PDAC. Serum RvD1 may provide an important diagnostic contribution in clinical practice to predict PDAC. Serum RvD1 levels were found to be predictive with high sensitivity and specificity in differentiating PDAC from metastatic PC. However, it was concluded that serum RvD1 levels cannot be used as a detection marker to differentiate PDAC from AP. RvD1 could be a representative agent of a new class of drugs to be proposed for innovative treatment of AP and PDAC. Our future study will investigate whether RvD1 can be a marker to differentiate from chronic pancreatitis.
Collapse
Affiliation(s)
- Yasemin Pekmezci
- Department of General Surgery, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey; (Y.P.); (S.E.); (B.C.T.); (S.P.); (M.V.)
| | - Sefa Ergun
- Department of General Surgery, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey; (Y.P.); (S.E.); (B.C.T.); (S.P.); (M.V.)
| | - Basar Can Turgut
- Department of General Surgery, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey; (Y.P.); (S.E.); (B.C.T.); (S.P.); (M.V.)
| | - Seyma Dumur
- Department of Medical Biochemistry, Faculty of Medicine, İstanbul Atlas University, Istanbul 34403, Turkey;
| | - Ugurcan Sayili
- Department of Public Health, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey;
| | - Hafize Uzun
- Department of Medical Biochemistry, Faculty of Medicine, İstanbul Atlas University, Istanbul 34403, Turkey;
| | - Salih Pekmezci
- Department of General Surgery, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey; (Y.P.); (S.E.); (B.C.T.); (S.P.); (M.V.)
| | - Mehmet Velidedeoglu
- Department of General Surgery, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34098, Turkey; (Y.P.); (S.E.); (B.C.T.); (S.P.); (M.V.)
| |
Collapse
|
38
|
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
|
39
|
Gálvez‐Montosa F, Peduzzi G, Sanchez‐Maldonado JM, ter Horst R, Cabrera‐Serrano AJ, Gentiluomo M, Macauda A, Luque N, Ünal P, García‐Verdejo FJ, Li Y, López López JA, Stein A, Bueno‐de‐Mesquita HB, Arcidiacono PG, Zanette DL, Kahlert C, Perri F, Soucek P, Talar‐Wojnarowska R, Theodoropoulos GE, Izbicki JR, Tamás H, Van Laarhoven H, Nappo G, Petrone MC, Lovecek M, Vermeulen RCH, Adamonis K, Reyes‐Zurita FJ, Holleczek B, Sumskiene J, Mohelníková‐Duchoňová B, Lawlor RT, Pezzilli R, Aoki MN, Pasquali C, Petrenkiene V, Basso D, Bunduc S, Comandatore A, Brenner H, Ermini S, Vanella G, Goetz MR, Archibugi L, Lucchesi M, Uzunoglu FG, Busch O, Milanetto AC, Puzzono M, Kupcinskas J, Morelli L, Sperti C, Carrara S, Capurso G, van Eijck CHJ, Oliverius M, Roth S, Tavano F, Kaaks R, Szentesi A, Vodickova L, Luchini C, Schöttker B, Landi S, Dohan O, Tacelli M, Greenhalf W, Gazouli M, Neoptolemos JP, Cavestro GM, Boggi U, Latiano A, Hegyi P, Ginocchi L, Netea MG, Sánchez‐Rovira P, Canzian F, Campa D, Sainz J. Polymorphisms within autophagy-related genes as susceptibility biomarkers for pancreatic cancer: A meta-analysis of three large European cohorts and functional characterization. Int J Cancer 2025; 156:339-352. [PMID: 39319538 PMCID: PMC11578083 DOI: 10.1002/ijc.35196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/17/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers with patients having unresectable or metastatic disease at diagnosis, with poor prognosis and very short survival. Given that genetic variation within autophagy-related genes influences autophagic flux and susceptibility to solid cancers, we decided to investigate whether 55,583 single nucleotide polymorphisms (SNPs) within 234 autophagy-related genes could influence the risk of developing PDAC in three large independent cohorts of European ancestry including 12,754 PDAC cases and 324,926 controls. The meta-analysis of these populations identified, for the first time, the association of the BIDrs9604789 variant with an increased risk of developing the disease (ORMeta = 1.31, p = 9.67 × 10-6). We also confirmed the association of TP63rs1515496 and TP63rs35389543 variants with PDAC risk (OR = 0.89, p = 6.27 × 10-8 and OR = 1.16, p = 2.74 × 10-5). Although it is known that BID induces autophagy and TP63 promotes cell growth, cell motility and invasion, we also found that carriers of the TP63rs1515496G allele had increased numbers of FOXP3+ Helios+ T regulatory cells and CD45RA+ T regulatory cells (p = 7.67 × 10-4 and p = 1.56 × 10-3), but also decreased levels of CD4+ T regulatory cells (p = 7.86 × 10-4). These results were in agreement with research suggesting that the TP63rs1515496 variant alters binding sites for FOXA1 and CTCF, which are transcription factors involved in modulating specific subsets of regulatory T cells. In conclusion, this study identifies BID as new susceptibility locus for PDAC and confirms previous studies suggesting that the TP63 gene is involved in the development of PDAC. This study also suggests new pathogenic mechanisms of the TP63 locus in PDAC.
Collapse
Affiliation(s)
| | | | - José Manuel Sanchez‐Maldonado
- Department of Biochemistry and Molecular Biology IUniversity of GranadaGranadaSpain
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTSGranadaSpain
- Instituto de Investigación Biosanataria Ibs.GranadaGranadaSpain
- Genomic Epidemiology GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Rob ter Horst
- Department of Internal Medicine and Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenThe Netherlands
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Antonio J. Cabrera‐Serrano
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTSGranadaSpain
- Instituto de Investigación Biosanataria Ibs.GranadaGranadaSpain
| | | | - Angelica Macauda
- Genomic Epidemiology GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Natalia Luque
- Department of Medical OncologyComplejo Hospitalario de JaénJaénSpain
| | - Pelin Ünal
- Genomic Epidemiology GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Yang Li
- Department of Internal Medicine and Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenThe Netherlands
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | | | - Angelika Stein
- Genomic Epidemiology GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Paolo Giorgio Arcidiacono
- Pancreatico/Biliary Endoscopy & Endosonography Division, Pancreas Translational & Clinical Research CenterSan Raffaele Scientific InstituteMilanItaly
| | - Dalila Luciola Zanette
- Laboratory for Applied Science and Technology in HealthCarlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz)CuritibaBrazil
| | - Christoph Kahlert
- Department of General SurgeryUniversity of HeidelbergHeidelbergBaden‐WürttembergGermany
| | - Francesco Perri
- Division of Gastroenterology and Research LaboratoryFondazione IRCCS “Casa Sollievo della Sofferenza” HospitalFoggiaItaly
| | - Pavel Soucek
- Biomedical Center, Faculty of Medicine in PilsenCharles UniversityPilsenCzech Republic
| | | | - George E. Theodoropoulos
- Colorectal Unit, First Department of Propaedeutic SurgeryMedical School of National and Kapodistrian University of Athens, Hippocration General HospitalAthensGreece
| | - Jakob R. Izbicki
- Department of General, Visceral and Thoracic SurgeryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Hussein Tamás
- Center for Translational MedicineSemmelweis UniversityBudapestHungary
- Division of Pancreatic Diseases, Heart and Vascular CenterSemmelweis UniversityBudapestHungary
| | - Hanneke Van Laarhoven
- Department of Medical OncologyAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Cancer Center AmsterdamImaging and BiomarkersAmsterdamThe Netherlands
| | - Gennaro Nappo
- Pancreatic UnitIRCCS Humanitas Research HospitalMilanItaly
- Department of Biomedical SciencesHumanitas UniversityMilanItaly
| | - Maria Chiara Petrone
- Pancreatico/Biliary Endoscopy & Endosonography Division, Pancreas Translational & Clinical Research CenterSan Raffaele Scientific InstituteMilanItaly
| | - Martin Lovecek
- Department of Surgery IUniversity Hospital OlomoucOlomoucCzech Republic
| | | | - Kestutis Adamonis
- Gastroenterology Department and Institute for Digestive ResearchLithuanian University of Health SciencesKaunasLithuania
| | | | - Bernd Holleczek
- Saarland Cancer RegistrySaarbrückenGermany
- Division of Clinical Epidemiology and Aging ResearchGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Jolanta Sumskiene
- Gastroenterology Department and Institute for Digestive ResearchLithuanian University of Health SciencesKaunasLithuania
| | | | - Rita T. Lawlor
- ARC‐Net Centre for Applied Research on Cancer University of VeronaVeronaItaly
- Department of Diagnostics and Public Health, Section of PathologyUniversity of VeronaVeronaItaly
| | | | - Mateus Nobrega Aoki
- Laboratory for Applied Science and Technology in HealthCarlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz)CuritibaBrazil
| | | | - Vitalija Petrenkiene
- Gastroenterology Department and Institute for Digestive ResearchLithuanian University of Health SciencesKaunasLithuania
| | - Daniela Basso
- Department of DIMEDLaboratory Medicine, University of PadovaPadovaItaly
| | - Stefania Bunduc
- Center for Translational MedicineSemmelweis UniversityBudapestHungary
- Division of Pancreatic Diseases, Heart and Vascular CenterSemmelweis UniversityBudapestHungary
- Carol Davila University of Medicine and PharmacyBucharestRomania
- Digestive Diseases and Liver Transplantation CenterFundeni Clinical InstituteBucharestRomania
| | - Annalisa Comandatore
- General Surgery Unit, Department of Translational Research and New Technologies in MedicineUniversity of PisaPisaItaly
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging ResearchGerman Cancer Research Center (DKFZ)HeidelbergGermany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Giuseppe Vanella
- Digestive and Liver Disease UnitS Andrea HospitalRomeItaly
- Pancreas Translational and Clinical Research CenterPancreato‐Biliary Endoscopy and Endoscopic Ultrasound, San Raffaele Scientific Institute IRCCSMilanItaly
| | - Mara R. Goetz
- Department of General, Visceral and Thoracic SurgeryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Livia Archibugi
- Digestive and Liver Disease UnitS Andrea HospitalRomeItaly
- Pancreas Translational and Clinical Research CenterPancreato‐Biliary Endoscopy and Endoscopic Ultrasound, San Raffaele Scientific Institute IRCCSMilanItaly
| | | | - Faik Guntac Uzunoglu
- Department of General, Visceral and Thoracic SurgeryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Olivier Busch
- Cancer Center AmsterdamImaging and BiomarkersAmsterdamThe Netherlands
- Department of Medical OncologyAmsterdam UMC Location University of AmsterdamAmsterdamThe Netherlands
| | | | - Marta Puzzono
- Gastroenterology and Gastrointestinal Endoscopy UnitVita‐Salute San Raffaele University, IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Juozas Kupcinskas
- Gastroenterology Department and Institute for Digestive ResearchLithuanian University of Health SciencesKaunasLithuania
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in MedicineUniversity of PisaPisaItaly
| | | | - Silvia Carrara
- Department of GastroenterologyIRCCS Humanitas Research Hospital – Endoscopic UnitMilanItaly
| | - Gabriele Capurso
- Digestive and Liver Disease UnitS Andrea HospitalRomeItaly
- Pancreas Translational and Clinical Research CenterPancreato‐Biliary Endoscopy and Endoscopic Ultrasound, San Raffaele Scientific Institute IRCCSMilanItaly
| | | | - Martin Oliverius
- Department of Surgery, University Hospital Kralovske Vinohrady, Third Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Susanne Roth
- Department of General SurgeryUniversity of HeidelbergHeidelbergBaden‐WürttembergGermany
| | - Francesca Tavano
- Division of Gastroenterology and Research LaboratoryFondazione IRCCS “Casa Sollievo della Sofferenza” HospitalFoggiaItaly
| | - Rudolf Kaaks
- Division of Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Andrea Szentesi
- Institute for Translational Medicine, Medical SchoolUniversity of PécsPécsHungary
| | - Ludmila Vodickova
- Department of Molecular Biology of Cancer, Institute of Experimental MedicineCzech Academy of SciencesPragueCzech Republic
- Institute of Biology and Medical Genetics, First Faculty of MedicineCharles UniversityPragueCzech Republic
- Faculty of Medicine and Biomedical Center in PilsenCharles UniversityPilsenCzech Republic
| | - Claudio Luchini
- ARC‐Net Centre for Applied Research on Cancer University of VeronaVeronaItaly
- Department of Engineering for Innovation in MedicineUniversity of VeronaVeronaItaly
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging ResearchGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Orsolya Dohan
- Division of Pancreatic Diseases, Heart and Vascular CenterSemmelweis UniversityBudapestHungary
| | - Matteo Tacelli
- Pancreatico/Biliary Endoscopy & Endosonography Division, Pancreas Translational & Clinical Research CenterSan Raffaele Scientific InstituteMilanItaly
| | - William Greenhalf
- Institute for Health Research Liverpool Pancreas Biomedical Research UnitUniversity of LiverpoolLiverpoolUK
| | - Maria Gazouli
- Department of Basic Medical Science, Laboratory of Biology, Medical SchoolNational and Kapodistrian University of AthensAthensGreece
| | - John P. Neoptolemos
- Department of General SurgeryUniversity of HeidelbergHeidelbergBaden‐WürttembergGermany
| | - Giulia Martina Cavestro
- Gastroenterology and Gastrointestinal Endoscopy UnitVita‐Salute San Raffaele University, IRCCS San Raffaele Scientific InstituteMilanItaly
| | - Ugo Boggi
- Division of General and Transplant SurgeryPisa University HospitalPisaItaly
| | - Anna Latiano
- Division of Gastroenterology and Research LaboratoryFondazione IRCCS “Casa Sollievo della Sofferenza” HospitalFoggiaItaly
| | - Péter Hegyi
- Center for Translational MedicineSemmelweis UniversityBudapestHungary
- Division of Pancreatic Diseases, Heart and Vascular CenterSemmelweis UniversityBudapestHungary
- Institute for Translational Medicine, Medical SchoolUniversity of PécsPécsHungary
- János Szentágothai Research CenterUniversity of PécsPécsHungary
| | - Laura Ginocchi
- Oncologia Massa CarraraAzienda USL Toscana Nord OvestCarraraItaly
| | - Mihai G. Netea
- Centre for Individualised Infection Medicine (CiiM) & TWINCOREjoint Ventures Between the Helmholtz‐Centre for Infection Research (HZI) and the Hannover Medical School (MHH)HannoverGermany
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES)University of BonnBonnGermany
| | | | - Federico Canzian
- Genomic Epidemiology GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Juan Sainz
- Department of Biochemistry and Molecular Biology IUniversity of GranadaGranadaSpain
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTSGranadaSpain
- Instituto de Investigación Biosanataria Ibs.GranadaGranadaSpain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP)BarcelonaSpain
| |
Collapse
|
40
|
Verras GI, Hamady ZZ, Collins A, Tapper W. Utility of Polygenic Risk Scores (PRSs) in Predicting Pancreatic Cancer: A Systematic Review and Meta-Analysis of Common-Variant and Mixed Scores with Insights into Rare Variant Analysis. Cancers (Basel) 2025; 17:241. [PMID: 39858023 PMCID: PMC11764467 DOI: 10.3390/cancers17020241] [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: 11/26/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Pancreatic adenocarcinoma is the most common histological subtype of pancreatic cancer, representing approximately 85% of all cases [...].
Collapse
Affiliation(s)
- Georgios Ioannis Verras
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (Z.Z.H.); (A.C.)
- Department of General Surgery, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Zaed Z. Hamady
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (Z.Z.H.); (A.C.)
- Department of General Surgery, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Andrew Collins
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (Z.Z.H.); (A.C.)
| | - William Tapper
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (Z.Z.H.); (A.C.)
| |
Collapse
|
41
|
Onji M, Kozono S, Nakai A, Kakizoe S, Tatsuguchi T, Naito K. Association between preoperative exercise tolerance and unplanned readmission in patients who underwent pancreatectomy for pancreatic ductal adenocarcinoma: a retrospective analysis. BMC Cancer 2025; 25:77. [PMID: 39806285 PMCID: PMC11731558 DOI: 10.1186/s12885-025-13466-9] [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: 08/07/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Despite advances in treatment, the incidence of postoperative complications following pancreatectomy remains high, leading to frequent hospital readmissions. Therefore, this study aimed to investigate the relationship between preoperative exercise tolerance and the likelihood of unplanned readmission in patients with pancreatic ductal adenocarcinoma. METHODS This retrospective analysis included 88 patients who underwent pancreatectomy at a single institution between July 2019 and September 2022 and focused on patients with pancreatic ductal adenocarcinoma. Patients' preoperative exercise tolerance was assessed using the 6-minute walk distance (6 MWD). RESULTS The study found a 22.7% readmission rate within 1 year, with the median 6 MWD being significantly lower in readmitted patients than in those who were not readmitted (390.0 m versus 436.5 m; p < 0.01). A 6 MWD cut-off of 425 m was a strong predictor of readmission, with lower preoperative exercise tolerance associated with a higher risk of readmission. CONCLUSIONS Enhancing preoperative physical reserves through exercise therapy may reduce readmission rates and improve patient outcomes. Further research with larger sample sizes is required to confirm these findings.
Collapse
Affiliation(s)
- Makoto Onji
- Department of Rehabilitation, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, 802-0077, Japan.
| | - Shingo Kozono
- Department of Surgery, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, 802-0077, Japan
| | - Asuka Nakai
- Department of Rehabilitation, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, 802-0077, Japan
| | - Shinji Kakizoe
- Department of Rehabilitation, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, 802-0077, Japan
| | - Takaaki Tatsuguchi
- Department of Surgery, Kitakyushu Municipal Medical Center, 2-1-1 Bashaku, Kokurakita-ku, Kitakyushu, 802-0077, Japan
| | - Koichi Naito
- Department of Physical Therapy, Faculty of Medical Science, Nagoya Women's University, 3-4-0 Shioji-cho, Mizuho-ku, Nagoya, 467-8610, Aichi, Japan
| |
Collapse
|
42
|
Yen CH, Lin CJ, Chen PY, Chen YJ, Wei LR, Chen PH, Yeh YC, Wang LHC, Chang HS, Tsai WC. Taming Pancreatic Cancer: Ardisia virens Kurz-Derived 4-Hydroxy-2-Methoxy-6-Tridecylphenyl Acetate as a Potent Tubulin Polymerization Inhibitor for Targeted Pancreatic Ductal Adenocarcinoma Therapy. Int J Med Sci 2025; 22:651-661. [PMID: 39898251 PMCID: PMC11783067 DOI: 10.7150/ijms.104112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/21/2024] [Indexed: 02/04/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a major global health challenge owing to late diagnosis and inherently metastatic nature. Although surgical intervention offers a potential remedy, only few patients are eligible, and drug resistance further complicates treatment. The therapeutic limitations have catalyzed a search for alternative treatments, particularly natural products. High-throughput screening identified six extracts from the Ardisia genus, with four from Ardisia virens Kurz, and 4-hydroxy-2-methoxy-6-tridecylphenyl acetate (HMTA) as the most potent candidate. Herein, we explored the anti-cancer effects of HMTA on PDAC and found it induced strong cytotoxic effects on BxPC-3 and PANC-1 pancreatic cancer cell lines. HMTA inhibited cell proliferation and induced apoptosis, as evidenced by annexin V/PI labeling and caspase 3 activation. HMTA halted cancer cell proliferation at the G2/M phase and induced multinucleation. Molecular docking analysis revealed that HMTA potentially could interact with tubulin, and in vitro assay confirmed it suppresses tubulin polymerization. HMTA significantly inhibited BxPC-3 xenograft tumor growth in mice. Overall, these findings suggested that HMTA is a promising candidate for PDAC therapy.
Collapse
Affiliation(s)
- Chia-Hung Yen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chien-Ju Lin
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Peng-Yu Chen
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yi-Jin Chen
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
| | - Ling-Rung Wei
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Pei-Hsuan Chen
- Department of Biological Sciences, College of Science, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Yi-Chen Yeh
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Lily Hui-Ching Wang
- School of Medicine, National Tsing Hua University, Hsinchu 300, Taiwan
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Hsun-Shuo Chang
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wan-Chi Tsai
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| |
Collapse
|
43
|
Wehrle CJ, Chang J, Gross A, Perlmutter B, Naples R, Stackhouse K, Augustin T, Joyce D, Simon R, Schlegel A, Walsh RM, Naffouje SA, Parente A. Sequence of Chemotherapy May Not Impact Survival After Resection of Pancreatic Tail Adenocarcinoma. J Surg Oncol 2025. [DOI: 10.1002/jso.28086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 01/01/2025] [Indexed: 02/03/2025]
Abstract
ABSTRACTIntroductionPancreatic ductal adenocarcinoma (PDAC) of the body/tail is notably different than PDAC in the head of the pancreas. Surgery plus chemotherapy is known to improve outcomes for all PDAC. The sequence of this therapy is well studied in head cancers yet has never been evaluated systematically in relation to distal pancreatectomy (DP).MethodsPatients receiving DP for PDAC and who received chemotherapy were included. Patients were compared receiving neoadjuvant systemic therapy (NAST) only, adjuvant (AST) only, both NAST + AST, and who received total neoadjuvant therapy (TNT), defined as > 24 weeks NAST before DP. PSM was performed 1:1 between AST and each other group creating quadruplets of patients for analysis. Matching factors were determined by multivariate cox‐regression analysis of factors independently affecting survival. Survival was considered from diagnosis and from surgery to account for potential biases.ResultsIn total, 4677 patients were selected with 400 (8.6%) receiving TNT, 536 (11.5%) NAST, 3235 (69.2%) AST, and 506 (10.8%) NAST + AST. A total of 341 quadruplets were selected after PSM. There were no differences in comorbidities, T/N‐stage, retrieved or positive lymph nodes, and margin status after matching. Kaplan–Meier analysis showed no difference in median OS between the matched treatment groups (33.71 ± 2.07 vs. 35.22 ± 1.62 vs. 32.53 ± 3.31 vs. 37.88 ± 1.90, respectively; log‐rank p = 0.464). Five‐year OS was not different between the groups (21% vs. 18% vs. 20% vs. 25%, respectively; p = 0.501).ConclusionThe sequence of chemotherapy and surgery did not impact survival in distal PDAC. Providers should tailor an individualized approach designed to maximize the chance of completing both treatments.
Collapse
Affiliation(s)
- Chase J. Wehrle
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Jenny Chang
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Abby Gross
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Breanna Perlmutter
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Robert Naples
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | | | - Toms Augustin
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Daniel Joyce
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Robert Simon
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Andrea Schlegel
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - R. Matthew Walsh
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Samer A. Naffouje
- Department of General Surgery Cleveland Clinic Foundation Cleveland Ohio USA
| | - Alessandro Parente
- Institute of Liver Studies, King's College Hospital NHS Foundation Trust London UK
| |
Collapse
|
44
|
Li J, Xia C, Li Y, Liu H, Gong C, Liang D. Effects of NK cell-related lncRNA on the immune microenvironment and molecular subtyping for pancreatic ductal adenocarcinoma. Front Immunol 2025; 15:1514259. [PMID: 39872533 PMCID: PMC11770056 DOI: 10.3389/fimmu.2024.1514259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 12/11/2024] [Indexed: 01/30/2025] Open
Abstract
Background Patients with pancreatic ductal adenocarcinoma (PDAC) face a highly unfavorable outcome and have a poor response to standard treatments. Immunotherapy, especially therapy based on natural killer (NK) cells, presents a promising avenue for the treatment of PDAC. Aims This research endeavor seeks to formulate a predictive tool specifically designed for PDAC based on NK cell-related long non-coding RNA (lncRNA), revealing new molecular subtypes of PDAC to promote personalized and precision treatment. Methods Utilizing the Tumor Immune Single-cell Hub 2 platform, we discovered genes associated with NK cells in PDAC. We employed the TCGA-PAAD dataset to ascertain the expression profiles of these NK cell-related genes and to screen for lncRNAs correlated with NK cells. Subsequently, we utilized Cox regression analysis for hazard ratios and LASSO regression analysis to identify three NK cell-related lncRNAs that were used to develop a prognostic assessment model. The forecasting accuracy of this model was appraised using the ROC curve and validated using a test set and the complete dataset. Results Successful construction of a prognostic model comprising three lncRNAs was achieved, demonstrating good predictive efficiency in the training set, validation dataset, and the entire dataset. NK cells display robust interactions with malignant cells, CD8 T cells, and fibroblasts in the PDAC tumor microenvironment and participate in the transport of various signaling molecules and following immune responses in PDAC. According to the expression patterns of NK cell-related lncRNA, we labeled PDAC patients as four molecular subtypes, exhibiting significant differences in immune cell infiltration, drug sensitivity, and other aspects. Conclusion This study Uncovered the activity of NK cells within PDAC, proposed an NK cell-related lncRNA model, and delineated new molecular subtypes, thereby providing targets for personalized therapy.
Collapse
Affiliation(s)
- Jinze Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Chuqi Xia
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yuxuan Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hanhan Liu
- Department of Pathology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Gong
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Daoming Liang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
45
|
Ejie J, Ashraf Ganjouei A, Hernandez S, Wang JJ, Romero-Hernandez F, Foroutani L, Hirose K, Nakakura E, Corvera CU, Alseidi A, Adam MA. Ongoing Failure to Deliver Guideline-Concordant Care for Patients with Pancreatic Cancer. Cancers (Basel) 2025; 17:170. [PMID: 39857951 PMCID: PMC11763659 DOI: 10.3390/cancers17020170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/01/2025] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
(1) Background: Comprehensive evaluation of guideline-concordant care (GCC) across all PDAC stages has yet to be thoroughly conducted. This study aimed to characterize treatment patterns and assess factors associated with receiving GCC among patients with pancreatic ductal adenocarcinoma (PDAC) in California. (2) Methods: Data on adult patients with PDAC were extracted from the California Cancer Registry (2004-2020). GCC is defined according to the recommendations provided by the National Comprehensive Cancer Network. We used multivariable logistic regression to identify factors associated with receiving GCC. A Cox model was used to examine the association of GCC with overall survival. (3) Results: A total of 50,346 PDAC patients were included (stage 1: 10%; stage 2: 25%; stage 3: 11%; stage 4: 54%). Only 46.7% of all patients received GCC (stage 1: 20%; stage 2: 40%; stage 3: 69%; stage 4: 50%). Only 31% of stage 1 patients underwent surgery. Factors inversely associated with receiving GCC were Hispanic ethnicity (OR 0.78; p < 0.001), Black race (OR 0.74; p < 0.001), having no insurance (OR 0.40; p < 0.001]), and a Charlson-Deyo score of ≥2 (OR 0.68; p < 0.001). Adherence to GCC was associated with improved survival (Hazard Ratio 0.39; p < 0.001). Notably, patients with stage 1 PDAC who received GCC had a median survival of 47 months vs. 8 months for those who did not. (4) Conclusions: Although stage 1 PDAC patients have the greatest potential for survival with GCC, only 20% of patients received such treatment. Thus, it is crucial to identify and address the modifiable factors contributing to these suboptimal care patterns.
Collapse
Affiliation(s)
- Jonathan Ejie
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA; (J.E.); (S.H.); (J.J.W.); (F.R.-H.); (L.F.)
- School of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Amir Ashraf Ganjouei
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA; (J.E.); (S.H.); (J.J.W.); (F.R.-H.); (L.F.)
| | - Sophia Hernandez
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA; (J.E.); (S.H.); (J.J.W.); (F.R.-H.); (L.F.)
| | - Jaeyun Jane Wang
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA; (J.E.); (S.H.); (J.J.W.); (F.R.-H.); (L.F.)
| | - Fernanda Romero-Hernandez
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA; (J.E.); (S.H.); (J.J.W.); (F.R.-H.); (L.F.)
| | - Laleh Foroutani
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA; (J.E.); (S.H.); (J.J.W.); (F.R.-H.); (L.F.)
| | - Kenzo Hirose
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA; (J.E.); (S.H.); (J.J.W.); (F.R.-H.); (L.F.)
| | - Eric Nakakura
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA; (J.E.); (S.H.); (J.J.W.); (F.R.-H.); (L.F.)
| | - Carlos Uriel Corvera
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA; (J.E.); (S.H.); (J.J.W.); (F.R.-H.); (L.F.)
| | - Adnan Alseidi
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA; (J.E.); (S.H.); (J.J.W.); (F.R.-H.); (L.F.)
| | - Mohamed Abdelgadir Adam
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA; (J.E.); (S.H.); (J.J.W.); (F.R.-H.); (L.F.)
| |
Collapse
|
46
|
Gadwa J, Yu J, Piper M, Knitz MW, Darragh LB, Olimpo N, Corbo S, Beynor JI, Neupert B, Nguyen AT, Hodgson C, Nguyen D, Abdelazeem KN, Saviola A, Pousse L, Bransi A, Pincha M, Klein C, Amann M, Karam SD. Divergent response to radio-immunotherapy is defined by intrinsic features of the tumor microenvironment. J Immunother Cancer 2025; 13:e010405. [PMID: 39773568 PMCID: PMC11749720 DOI: 10.1136/jitc-2024-010405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Treatment with immunotherapy can elicit varying responses across cancer types, and the mechanistic underpinnings that contribute to response vrsus progression remain poorly understood. However, to date there are few preclinical models that accurately represent these disparate disease scenarios. METHODS Using combinatorial radio-immunotherapy consisting of PD-1 blockade, IL2Rβγ biased signaling, and OX40 agonism we were able to generate preclinical tumor models with conflicting responses, where head and neck squamous cell carcinoma (HNSCC) models respond and pancreatic ductal adenocarcinoma (PDAC) progresses. RESULTS By modeling these disparate states, we find that regulatory T cells (Tregs) are expanded in PDAC tumors undergoing treatment, constraining tumor reactive CD8 T cell activity. Consequently, the depletion of Tregs restores the therapeutic efficacy of our treatment and abrogates the disparity between models. Moreover, we show that through heterotopic implantations the site of tumor development defines the response to therapy, as implantation of HNSCC tumors into the pancreas resulted in comparable levels of tumor progression. CONCLUSIONS This work highlights the complexity of combining immunotherapies within the tumor microenvironment (TME) and further defines the immune and non-immune components of the TME as an intrinsic feature of immune suppression.
Collapse
Affiliation(s)
- Jacob Gadwa
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Justin Yu
- Department of Otolaryngology - Head & Neck Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Miles Piper
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Michael W Knitz
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Laurel B Darragh
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Nicholas Olimpo
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sophia Corbo
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jessica I Beynor
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Brooke Neupert
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Alexander T Nguyen
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chloe Hodgson
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Diemmy Nguyen
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Khalid Nm Abdelazeem
- Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Anthony Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | | - Ali Bransi
- Roche Innovation Centre Zurich, Schlieren, Switzerland
| | - Mudita Pincha
- Roche Innovation Centre Zurich, Schlieren, Switzerland
| | | | - Maria Amann
- Roche Innovation Centre Zurich, Schlieren, Switzerland
| | - Sana D Karam
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
47
|
Shrestha AK, Haas C. Pulmonary Embolism in Long Standing Diabetes: A Hint Towards Pancreatic Carcinoma. J Community Hosp Intern Med Perspect 2025; 15:118-122. [PMID: 39867139 PMCID: PMC11759090 DOI: 10.55729/2000-9666.1429] [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: 05/22/2024] [Revised: 09/30/2024] [Accepted: 10/15/2024] [Indexed: 01/28/2025] Open
Abstract
Pancreatic carcinoma has remained one of the leading causes of cancer-related mortality worldwide. Cancer originating in the head of pancreas is often detected early in the disease due to biliary obstruction resulting in jaundice. In contrast, cancer of the pancreatic body and tail remains indolent, presenting late with significantly increased tumor burden and distant metastasis. Unfortunately, a single laboratory screening study is neither sensitive nor specific for early detection of pancreatic cancer. In this report, we present a patient with longstanding diabetes incidentally detected to have pancreatic tail carcinoma while presenting with pulmonary embolism, emphasizing the need for pancreatic cancer screening studies in population with longstanding diabetes.
Collapse
Affiliation(s)
- Anish K Shrestha
- Medstar Health Internal Medicine Residency Program, Baltimore, MD, USA
| | | |
Collapse
|
48
|
Fithroni AB, Inoue H, Zhou S, Hakim TFN, Tada T, Suzuki M, Sakurai Y, Ishimoto M, Yamada N, Sauriasari R, Sauerwein WAG, Watanabe K, Ohtsuki T, Matsuura E. Novel Drug Delivery Particles Can Provide Dual Effects on Cancer "Theranostics" in Boron Neutron Capture Therapy. Cells 2025; 14:60. [PMID: 39791761 PMCID: PMC11719788 DOI: 10.3390/cells14010060] [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: 11/24/2024] [Revised: 12/18/2024] [Accepted: 12/27/2024] [Indexed: 01/12/2025] Open
Abstract
Boron (B) neutron capture therapy (BNCT) is a novel non-invasive targeted cancer therapy based on the nuclear capture reaction 10B (n, alpha) 7Li that enables the death of cancer cells without damaging neighboring normal cells. However, the development of clinically approved boron drugs remains challenging. We have previously reported on self-forming nanoparticles for drug delivery consisting of a biodegradable polymer, namely, "AB-type" Lactosome® nanoparticles (AB-Lac particles)- highly loaded with hydrophobic B compounds, namely o-Carborane (Carb) or 1,2-dihexyl-o-Carborane (diC6-Carb), and the latter (diC6-Carb) especially showed the "molecular glue" effect. Here we present in vivo and ex vivo studies with human pancreatic cancer (AsPC-1) cells to find therapeutically optimal formulas and the appropriate treatment conditions for these particles. The biodistribution of the particles was assessed by the tumor/normal tissue ratio (T/N) in terms of tumor/muscle (T/M) and tumor/blood (T/B) ratios using near-infrared fluorescence (NIRF) imaging with indocyanine green (ICG). The in vivo and ex vivo accumulation of B delivered by the injected AB-Lac particles in tumor lesions reached a maximum by 12 h post-injection. Irradiation studies conducted both in vitro and in vivo showed that AB-Lac particles-loaded with either 10B-Carb or 10B-diC6-Carb significantly inhibited the growth of AsPC-1 cancer cells or strongly inhibited their growth, with the latter method being significantly more effective. Surprisingly, a similar in vitro and in vivo irradiation study showed that ICG-labeled AB-Lac particles alone, i.e., without any 10B compounds, also revealed a significant inhibition. Therefore, we expect that our ICG-labeled AB-Lac particles-loaded with 10B compound(s) may be a novel and promising candidate for providing not only NIRF imaging for a practical diagnosis but also the dual therapeutic effects of induced cancer cell death, i.e., "theranostics".
Collapse
Affiliation(s)
- Abdul Basith Fithroni
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (A.B.F.); (H.I.); (S.Z.); (T.F.N.H.); (T.T.); (K.W.); (T.O.)
| | - Haruki Inoue
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (A.B.F.); (H.I.); (S.Z.); (T.F.N.H.); (T.T.); (K.W.); (T.O.)
| | - Shengli Zhou
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (A.B.F.); (H.I.); (S.Z.); (T.F.N.H.); (T.T.); (K.W.); (T.O.)
| | - Taufik Fatwa Nur Hakim
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (A.B.F.); (H.I.); (S.Z.); (T.F.N.H.); (T.T.); (K.W.); (T.O.)
| | - Takashi Tada
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (A.B.F.); (H.I.); (S.Z.); (T.F.N.H.); (T.T.); (K.W.); (T.O.)
| | - Minoru Suzuki
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka 590-0494, Japan; (M.S.); (Y.S.)
| | - Yoshinori Sakurai
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka 590-0494, Japan; (M.S.); (Y.S.)
| | | | - Naoyuki Yamada
- Nihon Fukushi Fuiin Holding, Co., Ltd., Fukushima 979-0513, Japan;
| | - Rani Sauriasari
- Faculty of Pharmacy, Universitas Indonesia, Depok 16424, Indonesia;
| | - Wolfgang A. G. Sauerwein
- Deutsche Gesellschaft für Bor-Neutroneneinfangtherapie DGBNCT e.V., University Hospital Essen, Klinik für Strahlentherapie, 45122 Essen, Germany;
| | - Kazunori Watanabe
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (A.B.F.); (H.I.); (S.Z.); (T.F.N.H.); (T.T.); (K.W.); (T.O.)
| | - Takashi Ohtsuki
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (A.B.F.); (H.I.); (S.Z.); (T.F.N.H.); (T.T.); (K.W.); (T.O.)
| | - Eiji Matsuura
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan; (A.B.F.); (H.I.); (S.Z.); (T.F.N.H.); (T.T.); (K.W.); (T.O.)
- Faculty of Pharmacy, Universitas Indonesia, Depok 16424, Indonesia;
- Collaborative Research Center for OMIC, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
- Neutron Therapy Research Center (NTRC), Okayama University, Okayama 700-8558, Japan
| |
Collapse
|
49
|
Elfekih H, Charfeddine Y, Said MA, Saafi W, Jaziri H, Mraidha MH, Halloul I, Ben Ali A, Yacoub S, Brahem S, Guesmi A, Ksiaa M, Ben Ahmed S, Sahli J, Saad G, Hasni Y. Comparative Analysis of Long-Standing and Newly Diagnosed Diabetes Mellitus in Patients with Pancreatic Ductal Adenocarcinoma: A Tunisian Multicenter Study. LA TUNISIE MEDICALE 2025; 103:112-116. [PMID: 39812203 PMCID: PMC11906228 DOI: 10.62438/tunismed.v103i1.5328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/23/2024] [Indexed: 01/16/2025]
Abstract
INTRODUCTION Diabetes mellitus has emerged as a global public health issue due to its increasing prevalence and the increased risk of developing cancers. Pancreatic cancer is believed to be both a consequence of pre-existing diabetes and a potential cause of new-onset diabetes. AIM This study aims to compare the characteristics of patients with pancreatic ductal adenocarcinoma and newly diagnosed or long-standing diabetes mellitus. METHODS A multicentric retrospective study was conducted over 13 years at two university hospitals in Sousse, Tunisia. Included patients had whether a newly diagnosed or a long-standing diabetes mellitus with histologically confirmed pancreatic ductal adenocarcinoma. Statistical analysis using appropriate tests was conducted. RESULTS The prevalence of diabetes mellitus was 44.6% among three hundred and seven patients with pancreatic cancer. The male-to-female ratio in patients with pancreatic ductal adenocarcinoma was 2.6:1. Patients' mean age was 63.9 years, with the majority being over 50 years old. Most patients had no family history of diabetes and exhibited significant weight loss, low body mass index, and uncontrolled diabetes. The comparison between individuals with newly diagnosed diabetes and those with long-standing diabetes revealed numerous similarities, apart from significant differences in drinking patterns (p = 0.03), tumor size (p = 0.018), and smoking in the subgroup of males (p = 0.044). CONCLUSION Patients over 50 with newly diagnosed diabetes mellitus, particularly those who consume alcohol occasionally and men who are not heavy smokers, should undergo further evaluation to identify potential early-stage pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Hamza Elfekih
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Endocrinology Diabetology, 4000, Sousse, Tunisia
| | - Yasmine Charfeddine
- University of Sousse, Faculty of Medicine of Sousse, Department of Family and Community Medicine, 4002, Sousse, Tunisia
| | - Mohamed Amine Said
- University of Sousse, Faculty of Medicine of Sousse, 4002, Sahloul University Hospital, Department of Surgery, 4054, Sousse, Tunisia
| | - Wiem Saafi
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Endocrinology Diabetology, 4000, Sousse, Tunisia
| | - Hanen Jaziri
- University of Sousse, Faculty of Medicine of Sousse, 4002, Sahloul University Hospital, Department of Gastroenterology, 4054, Sousse, Tunisia
| | - Mohamed Hedi Mraidha
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Surgery, 4000, Sousse, Tunisia
| | - Imen Halloul
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Endocrinology Diabetology, 4000, Sousse, Tunisia
| | - Azer Ben Ali
- University of Sousse, Faculty of Medicine of Sousse, 4002, Sahloul University Hospital, Department of Surgery, 4054, Sousse, Tunisia
| | - Sarra Yacoub
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Pathology, 4000, Sousse, Tunisia
| | - Salem Brahem
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Radiology, 4000, Sousse, Tunisia
| | - Ayoub Guesmi
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Radiology, 4000, Sousse, Tunisia
| | - Mehdi Ksiaa
- University of Sousse, Faculty of Medicine of Sousse, 4002, Sahloul University Hospital, Department of Gastroenterology, 4054, Sousse, Tunisia
| | - Slim Ben Ahmed
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Medical Oncology, 4000, Sousse, Tunisia
| | - Jihen Sahli
- University of Sousse, Faculty of Medicine of Sousse, Department of Family and Community Medicine, 4002, Sousse, Tunisia
| | - Ghada Saad
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Endocrinology Diabetology, 4000, Sousse, Tunisiaa
| | - Yosra Hasni
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Endocrinology Diabetology, 4000, Sousse, Tunisia
| |
Collapse
|
50
|
Adachi T, Nakamura M, Iwai T, Yoshimura M, Mizowaki T. Delta-Radiomics Approach Using Contrast-Enhanced and Noncontrast-Enhanced Computed Tomography Images for Predicting Distant Metastasis in Patients With Borderline Resectable Pancreatic Carcinoma. Adv Radiat Oncol 2025; 10:101669. [PMID: 39687476 PMCID: PMC11647472 DOI: 10.1016/j.adro.2024.101669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 10/21/2024] [Indexed: 12/18/2024] Open
Abstract
Purpose To predict distant metastasis (DM) in patients with borderline resectable pancreatic carcinoma using delta-radiomics features calculated from contrast-enhanced computed tomography (CECT) and non-CECT images. Methods and Materials Among 250 patients who underwent radiation therapy at our institution between February 2013 and December 2021, 67 patients were deemed eligible. A total of 11 clinical features and 3906 radiomics features were incorporated. Radiomics features were extracted from CECT and non-CECT images, and the differences between these features were calculated, resulting in delta-radiomics features. The patients were randomly divided into the training (70%) and test (30%) data sets for model development and validation. Predictive models were developed with clinical features (clinical model), radiomics features (radiomics model), and a combination of the abovementioned features (hybrid model) using Fine-Gray regression (FG) and random survival forest (RSF). Optimal hyperparameters were determined using stratified 5-fold cross-validation. Subsequently, the developed models were applied to the remaining test data sets, and the patients were divided into high- or low-risk groups based on their risk scores. Prognostic power was assessed using the concordance index, with 95% CIs obtained through 2000 bootstrapping iterations. Statistical significance between the above groups was assessed using Gray's test. Results At a median follow-up period of 23.8 months, 47 (70.1%) patients developed DM. The concordance indices of the FG-based clinical, radiomics, and hybrid models were 0.548, 0.603, and 0.623, respectively, in the test data set, whereas those of the RSF-based models were 0.598, 0.680, and 0.727, respectively. The RSF-based model, including delta-radiomics features, significantly divided the cumulative incidence curves into two risk groups (P < .05). The feature map of the gray-level size-zone matrix showed that the difference in feature values between CECT and non-CECT images correlated with the incidence of DM. Conclusions Delta-radiomics features obtained from CECT and non-CECT images using RSF successfully predict the incidence of DM in patients with borderline resectable pancreatic carcinoma.
Collapse
Affiliation(s)
- Takanori Adachi
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Mitsuhiro Nakamura
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
- Department of Advanced Medical Physics, Graduate School of Medicine, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Takahiro Iwai
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Michio Yoshimura
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Takashi Mizowaki
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| |
Collapse
|