1
|
Yan F, Yu L, Liu Z, Qi J, Wang L, Zhou M, Yin P. Subnational trend and driving factors for pancreatic cancer burden in China, 1990-2021: an analysis based on the Global Burden of Disease Study 2021. Ann Med 2025; 57:2484465. [PMID: 40172666 PMCID: PMC11966975 DOI: 10.1080/07853890.2025.2484465] [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: 10/22/2024] [Revised: 03/04/2025] [Accepted: 03/16/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND The mortality of pancreatic cancer in China showed an increasing trend between 2005 and 2020, with significant discrepancies in the burden of pancreatic cancer in provinces. METHODS We analyzed numbers of death, incidence, disability-adjusted life years (DALY) and corresponding age-standardized rates for pancreatic cancer in China using data from the Global Burden of Disease Study 2021. We conducted trend analysis in pancreatic cancer burden over time by age group and gender. Decomposition analysis was used to assess the drivers of change in cancer-related deaths in China due to three explanatory factors: population growth, population ageing and age-specific mortality. RESULTS In 2021, the ASMR of pancreatic cancer in China was 5.72/100,000(95%UI: 4.59, 6.91), the age-standardized incidence (ASIR) rate was 5.64/100,000(95%UI: 4.52, 6.84) and the age-standardized DALY rate was 137.23/100,000 (95%UI:108.15, 166.74). From 1990 to 2021, the ASMR of pancreatic cancer in China generally showed an increasing trend (AAPC: 0.56, 95%UI: 0.52, 0.59). The burden of pancreatic cancer was consistently higher in Chinese men compared to women during the study period.Compared with 1990, the number of deaths from pancreatic cancer has increased in all provinces of China in 2021, with the overall number of deaths increasing by 67.49%. Population ageing was the major cause of the increase in deaths from pancreatic cancer in China, accounting for 45.89%. CONCLUSIONS The burden of pancreatic cancer in China is still at a high level and population ageing is the main reason for the increase in pancreatic cancer deaths.
Collapse
Affiliation(s)
- Fanshu Yan
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Lingling Yu
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhe Liu
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jinlei Qi
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Lijun Wang
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Maigeng Zhou
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Peng Yin
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
2
|
Kobayashi K, Kishi Y, Tsunenari T, Yonamine N, Takao M, Einama T, Tsujimoto H, Ueno H. Dissecting Tumor Size Underestimation in Pancreatic Cancer: A Comparative Analysis of Preoperative Treatments. Ann Surg Oncol 2025; 32:3593-3602. [PMID: 39871078 PMCID: PMC11976789 DOI: 10.1245/s10434-025-16917-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/05/2025] [Indexed: 01/29/2025]
Abstract
BACKGROUND Tumor size (TS) in pancreatic ductal adenocarcinoma (PDAC) is one of the most important prognostic factors. However, discrepancies between TS on preoperative images (TSi) and pathological specimens (TSp) have been reported. This study aims to evaluate the factors associated with the differences between TSi and TSp. PATIENTS AND METHODS We retrospectively analyzed patients with PDAC who underwent surgery at our institution between January 2010 and November 2023. TS discrepancy (TSD[%]) was defined as ([TSp - TSi]/TSp) × 100. Using logistic regression, we generated a receiver operating characteristic (ROC) curve to define the cutoff for TSi underestimation predicting clinical tumor (T) stage migration. Univariate and multivariate analyses were performed to evaluate predictors of TSi underestimation. RESULTS Of the 231 patients, 99 (42%) patients received preoperative chemotherapy. The ROC curve determined a TSD underestimation cutoff of 25.9%. The number of TSp > TSi cases was 185 (80%), and TSi underestimation was present in 117 (51%) patients. T stage migration rates were 76%, 26%, and 50% in clinical stage (c) T1, cT2, cT3, respectively, among the patients with chemotherapy, and 93%, 33%, and 14%, respectively, in those without chemotherapy. Multivariate analyses revealed that independent predictors of TSi underestimation were posterior surface invasion in the patients with preoperative chemotherapy and anterior surface invasion in those without chemotherapy. CONCLUSIONS TS was more commonly underestimated than overestimated, and cT1 rarely corresponded to pathological (p)T1. The factors contributing to TSi underestimation differed between patients with and without preoperative chemotherapy. Therefore, these two groups should be considered separately for accurate TSi evaluation.
Collapse
Affiliation(s)
- Kazuki Kobayashi
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Yoji Kishi
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan.
| | - Takazumi Tsunenari
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Naoto Yonamine
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Mikiya Takao
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Takahiro Einama
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hironori Tsujimoto
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|
3
|
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
|
4
|
Hildebrand S, Pfeifer A. The obesity pandemic and its impact on non-communicable disease burden. Pflugers Arch 2025; 477:657-668. [PMID: 39924587 PMCID: PMC12003543 DOI: 10.1007/s00424-025-03066-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 02/11/2025]
Abstract
The rising prevalence of overweight and obesity across the globe is a major threat both to public health and economic development. This is mainly due to the link of obesity with the development and outcomes of non-communicable diseases (NCDs). NCDs are a leading cause of global death and disability, and reducing the burden of NCDs on patients and healthcare systems is of critical importance to improve public health. Obesity is projected to be the number one preventable risk factor for NCDs by 2035, and there is an urgent need to tackle the growing obesity rates in order to reduce NCD incidence and severity. Here, we review the current understanding of the impact of obesity on NCD burden in general, as well as the epidemiological and mechanistic relationship between obesity and some of the most common classes of NCDs. By literature review, we found that over 70% of NCDs have a documented association with obesity, highlighting the importance of a better understanding of the pathophysiologies underlying obesity/overweight as well as the interaction between obesity and NCDs in order to reduce global disease burden.
Collapse
Affiliation(s)
- Staffan Hildebrand
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, 53127, Bonn, Germany.
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, 53127, Bonn, Germany.
- PharmaCenter Bonn, University of Bonn, Bonn, Germany.
| |
Collapse
|
5
|
Zhou J, Yang K, Lu M, Fu P, Chen Y, Chen L. Higher density of compact B cell clusters in invasive front may contribute to better prognosis in pancreatic ductal adenocarcinoma. Discov Oncol 2025; 16:555. [PMID: 40246809 PMCID: PMC12006623 DOI: 10.1007/s12672-025-02260-5] [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: 10/07/2024] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
While the correlation between T cells and patient survival was widely investigated, the clinical significance of CD20+ B cells in pancreatic ductal adenocarcinoma (PDAC) is less clear. We hypothesized that the spatial pattern of B cells within tumor microenvironment (TME) are more informative, which may reveal the prognostic significance for PDAC patients. Therefore, we developed a computer-based workflow to analyze CD20+ B cells in whole slide images (WSI) from 45 cases of PDAC patients. Depending on this workflow, annotations of each case which were created by pathologists were subdivided for three regions, including invasive front (IF), cancer center (CT) and cancer island (CI) to explore the association between the spatial pattern of CD20+ B cells and patient prognosis outcomes. After that, occupancy rate (as area under curve, occupancy AUC), fractal dimension differences (ΔFD), cluster density and coverage ratio were used to quantify the spatial pattern of B cells in TME. We observed B cells were distributed across different regions, manifesting in both clustered and dispersed patterns. Compared to features of B cells spatial distribution in CT region, B cells in IF region exhibited higher occupancy AUC (p = 0.00004), cluster density (p = 0.000002) and coverage ratio (p = 0.000884). Patients with longer survivals had smaller ΔFD (p = 0.05), higher B-cell cluster density (p = 0.003) and lower coverage ratio (p = 0.02) in IF region. Our study indicated the spatial distribution of B cells in IF and CT was different and the higher density of compact B-cell clusters in IF region may be associated with better prognosis in PDAC.
Collapse
Affiliation(s)
- Junwen Zhou
- Department of Land Surveying and Geo-Informatics, The Hong Kong Polytechnic University, Hung Hom, HK, China
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Kunping Yang
- College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, China
| | - Mei Lu
- Fuqing City Hospital Affiliated to Fujian Medical University, Fuqing, Fujian, China
| | - Peiling Fu
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Yupeng Chen
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Linying Chen
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China.
| |
Collapse
|
6
|
Liu W, Rao L, Qiao Z, Wang G, Li B, Shen G. Global disparities in the burden of pancreatic cancer (1990-2021): insights from the 2021 Global Burden of Disease study. BMC Cancer 2025; 25:722. [PMID: 40247202 PMCID: PMC12007340 DOI: 10.1186/s12885-025-14110-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is a highly lethal malignancy, ranking seventh among cancer-related deaths worldwide. This study utilizes data from the 2021 Global Burden of Disease (GBD) study to examine the global burden of PC and associated health inequalities from 1990 to 2021, with a focus on key risk factors such as obesity, high fasting plasma glucose, and the Socio-Demographic Index (SDI). METHODS Disability-Adjusted Life Years (DALYs) for PC were estimated using GBD 2021 data. The analysis incorporated SDI, age, gender, and major risk factors, including obesity and high fasting plasma glucose. Descriptive statistics and visualizations, such as age-sex pyramids and geographic maps, were employed to assess global, regional, and national burdens. Health disparities were quantified using the Concentration Index (CI) and the Slope Index of Inequality (SII), with CI assessing relative health distribution by income and SII measuring absolute socioeconomic inequality. RESULTS Globally, PC-related DALYs rose from 1.76 million in 1990 to 4.25 million in 2021 (141.48% increase), with the age-standardized DALY rate up 11.57% to 48.71 (95% UI 23.43 to 74.33). The burden was highest in high SDI regions, while low SDI areas still faced elevated rates; transitional and developing economies showed the highest age-standardized DALY rates. The SII increased from 189.63 (95% CI 177.65 to 245.17) in 1990 to 321.17 (95% CI 294.48 to 379.722) in 2021, indicating widening socioeconomic disparities. CONCLUSION PC remains a significant global health challenge with growing socioeconomic and geographic disparities. Urgent action is needed to address modifiable risk factors (e.g., obesity, diabetes) through enhanced healthcare infrastructure, early detection, and treatment access in low SDI countries, alongside improved data systems and international collaboration.
Collapse
Affiliation(s)
- Wei Liu
- Department of Minimally Invasive Common Surgery, Suzhou Ninth People's Hospital, Xuzhou Medical University Suzhou Bay Clinical College, Suzhou, Jiangsu Province, China
| | - Li Rao
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhengguo Qiao
- Department of Gastroenterology, Suzhou Ninth People's Hospital, Xuzhou Medical University, Suzhou Bay Clinical College, Suzhou, Jiangsu Province, China
| | - Gang Wang
- Department of Minimally Invasive Common Surgery, Suzhou Ninth People's Hospital, Xuzhou Medical University Suzhou Bay Clinical College, Suzhou, Jiangsu Province, China
| | - Bin Li
- Department of Minimally Invasive Common Surgery, Suzhou Ninth People's Hospital, Xuzhou Medical University Suzhou Bay Clinical College, Suzhou, Jiangsu Province, China
| | - Genhai Shen
- Department of Minimally Invasive Common Surgery, Suzhou Ninth People's Hospital, Xuzhou Medical University Suzhou Bay Clinical College, Suzhou, Jiangsu Province, China.
| |
Collapse
|
7
|
Noguchi A, Kimura M, Saiki Y, Ishikawa T, Kokumai T, Omori Y, Ono Y, Miszukami Y, Ishida M, Mizuma M, Nakagawa K, Unno M, Furukawa T. Transcriptional regulation of ETV5 by mitogen-activated protein kinase via ETS-1 in human pancreatic cancer cells. Sci Rep 2025; 15:12128. [PMID: 40204967 PMCID: PMC11982384 DOI: 10.1038/s41598-025-97166-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 04/02/2025] [Indexed: 04/11/2025] Open
Abstract
Pancreatic cancer is characterized by constitutive activation of mitogen-activated protein kinase /extracellular signal-regulated kinase 1/2 (ERK1/2) driven by gain-of-function mutations of KRAS. Our previous transcriptome sequencing of ERK1/2-attenuated cultured pancreatic cancer cells unveiled numerous downstream genes activated by ERK1/2 including ETV5. In this study, we explored the mechanism of transcriptional regulation of ETV5 by ERK1/2 in human pancreatic cancer cells. Detailed reporter assays uncovered a core promoter region spanning between - 350 and - 985 from the transcription start site of ETV5 as a strong responsive element to ERK1/2 activity. Moreover, ETS proto-oncogene 1, transcription factor (ETS-1) was found to bind to one of consensus binding sites in the core region and to promote ERK1/2-mediated upregulation of ETV5. Investigation of functional significances of ETS variant transcription factor 5 (ETV5) expression in the pancreatic cancer cells revealed that ETV5 was associated with resistance to gemcitabine; while no significance in proliferation, migration, and invasion. ETV5 expression in pancreatic ductal adenocarcinoma tissues resected from patients undergoing neoadjuvant chemotherapy was associated with KRAS mutations, which was consistent with ETV5 as a downstream upregulated molecule of RAS-ERK1/2 pathway. This study elucidated the mechanism of ERK1/2-mediated transcriptional regulation of ETV5 in human cancer cells, which could contribute to understand pancreatic cancer pathobiology.
Collapse
Affiliation(s)
- Aya Noguchi
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aobaku, Sendai, 980-8575, Japan
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Masanobu Kimura
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aobaku, Sendai, 980-8575, Japan
| | - Yuriko Saiki
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aobaku, Sendai, 980-8575, Japan
| | - Tomohiko Ishikawa
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aobaku, Sendai, 980-8575, Japan
| | - Takashi Kokumai
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Yuko Omori
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aobaku, Sendai, 980-8575, Japan
| | - Yusuke Ono
- Institute of Biomedical Research, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Yusuke Miszukami
- Institute of Biomedical Research, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan
- Division of Gastroenterology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Masaharu Ishida
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Masamichi Mizuma
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Kei Nakagawa
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Toru Furukawa
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aobaku, Sendai, 980-8575, Japan.
| |
Collapse
|
8
|
Ulus G, Özbek EN, Yılmaz H, Keselik E, Sarıcaoğlu M, Akyol Bahçeci S, İşel E, Debeleç Bütüner B, Yetik Anacak G, Koparal AT. Borax pentahydrate as a promising boron-based angiogenesis inhibitor. J Trace Elem Med Biol 2025; 89:127640. [PMID: 40203787 DOI: 10.1016/j.jtemb.2025.127640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Boron, a trace element, is involved in various physiological and metabolic processes, and recent studies suggest that boron compounds may have potential in cancer prevention and treatment. In this study, the antiangiogenic effects of a boron compound, borax pentahydrate (BPH), were investigated. Angiogenesis is a tightly regulated biological process responsible for the formation of new blood vessels from existing vasculatures. This process plays a critical role in cancer progression, making it an important target for cancer therapy. Pancreatic and kidney cancers are difficult to treat because they are aggressive and resistant to chemotherapy. METHODS The antiproliferative activity was evaluated using the MTT assay, while antiangiogenic effects were tested through in vitro tube formation assays and in ovo chick chorioallantoic membrane (CAM) assay. The effect of BPH on VEGF levels was determined using Western blot analysis in HUVEC, ACHN, PANC-1 cells. The effect of BPH on tumor angiogenesis was investigated with an in vivo Ehrlich ascites carcinoma model (EAC). RESULTS BPH exhibited potent antiproliferative and antiangiogenic activities, inhibiting the proliferation of ACHN, PANC-1, and HUVECs, disrupting endothelial tube formation, and inhibiting vascular formation on the CAM surface in a dose-dependent manner. VEGF levels were significantly decreased in ACHN, PANC-1 and HUVECs. There was also a decrease in VEGF and TGF-β1 levels in BPH-treated tumor groups. In addition, BPH caused a decrease in tumor size. CONCLUSION These findings suggest that BPH may be a new antiangiogenic and antitumoral agent. BPH may contribute to drug development studies targeting angiogenesis-related diseases as a promising new therapeutic agent.
Collapse
Affiliation(s)
- G Ulus
- Republic of Türkiye, Ministry of Education, Şerife Bacı Vocational and Technical High School, Izmir 35090, Turkiye.
| | - E N Özbek
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkiye
| | - H Yılmaz
- Republic of Türkiye, Ministry of Education, Mimar Sinan Vocational and Technical High School, Izmir 35090, Turkiye
| | - E Keselik
- Department of Histology and Embryology, Faculty of Medicine, Katip Çelebi University, Izmir 35100, Turkiye
| | - M Sarıcaoğlu
- Department of Histology and Embryology, Faculty of Medicine, Katip Çelebi University, Izmir 35100, Turkiye
| | - S Akyol Bahçeci
- Department of Histology and Embryology, Faculty of Medicine, Katip Çelebi University, Izmir 35100, Turkiye
| | - E İşel
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkiye
| | - B Debeleç Bütüner
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkiye
| | - G Yetik Anacak
- Department of Pharmacology, Faculty of Pharmacy, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkiye
| | - A T Koparal
- Yunus Emre Vocational School of Health Services, Anadolu University, Eskisehir 26470, Turkiye
| |
Collapse
|
9
|
Li YC, Zhang L, Wang YT, Hu H, Zhang ZY, Nie QQ, Zuo CJ. Role of EFNAs in Shaping the Tumor Immune Microenvironment and Their Impact on Pancreatic Adenocarcinoma Prognosis. Cancer Manag Res 2025; 17:693-712. [PMID: 40190415 PMCID: PMC11972607 DOI: 10.2147/cmar.s502401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
Purpose Due to the highly heterogeneous and immunosuppressed tumor microenvironment (TME), pancreatic adenocarcinoma (PAAD) has limited therapeutic options and an abysmal prognosis. Ephrin A 1-5 (EFNA1-5) have been shown to regulate tumorigenesis and metastasis in various cancers, but its role in PAAD remains unclear. Methods We comprehensively analyzed EFNA gene expression levels in pan-cancer and PAAD using the GEPIA and HPA databases. Then, we assessed the prognostic value of EFNA1-5 using the Kaplan-Meier plotter and nomogram model. Further exploration of the association of EFNA1-5 with clinicopathological features of PAAD used information from the UALCAN database, and the TIMER dataset was used to reveal the correlation between EFNA1-5 and the tumor immune microenvironment (TIME) of pancreatic cancer. In addition, cBioPortal Databases, GSEA, and GSCALite were used to explore gene changes, protein interactions, and biological functions. Finally, the oncogenic effect of EFNA5 was verified in vivo and in vitro. Results The expression levels of EFNA1-5 were significantly upregulated in PAAD. The expression of EFNA1/3/4/5 were significantly associated with overall survival (OS) and relapse-free survival (RFS) in PAAD patients. The high expression of EFNA2-5 were related to poor clinical features, such as higher tumor stage or grade and a wider range of lymph node metastasis. EFNA1-5 were closely associated with immune cell infiltration, CAFs, and MDSCs expression. Furthermore, EFNA5 is an independent risk factor for poor prognosis in PAAD patients, and it can promote the malignant progression of pancreatic cancer in vitro and in vivo. Conclusion Differential expression of EFNA1-5 is associated with TIME in pancreatic cancer, predicts different survival outcomes, and maybe a novel prognostic marker reflecting an immunosuppressive state and a potential therapeutic target.
Collapse
Affiliation(s)
- Yu-Chao Li
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Lu Zhang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Yi-Ting Wang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Hao Hu
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Ze-Yu Zhang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Qian-Qian Nie
- Department of Central Laboratory, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Chang-Jing Zuo
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
10
|
Devasahayam Arokia Balaya R, Sen P, Grant CW, Zenka R, Sappani M, Lakshmanan J, Athreya AP, Kandasamy RK, Pandey A, Byeon SK. An integrative multi-omics analysis reveals a multi-analyte signature of pancreatic ductal adenocarcinoma in serum. J Gastroenterol 2025; 60:496-511. [PMID: 39666045 DOI: 10.1007/s00535-024-02197-6] [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: 02/16/2024] [Accepted: 12/01/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains a formidable health challenge due to its detection at a late stage and a lack of reliable biomarkers for early detection. Although levels of carbohydrate antigen 19-9 are often used in conjunction with imaging-based tests to aid in the diagnosis of PDAC, there is still a need for more sensitive and specific biomarkers for early detection of PDAC. METHODS We obtained serum samples from 88 subjects (patients with PDAC (n = 58) and controls (n = 30)). We carried out a multi-omics analysis to measure cytokines and related proteins using proximity extension technology and lipidomics and metabolomics using tandem mass spectrometry. Statistical analysis was carried out to find molecular alterations in patients with PDAC and a machine learning model was used to derive a molecular signature of PDAC. RESULTS We quantified 1,462 circulatory proteins along with 873 lipids and 1,001 metabolites. A total of 505 proteins, 186 metabolites and 33 lipids including bone marrow stromal antigen 2 (BST2), keratin 18 (KRT18), and cholesteryl ester(20:5) were found to be significantly altered in patients. We identified different levels of sphingosine, sphinganine, urobilinogen and lactose indicating that glycosphingolipid and galactose metabolisms were significantly altered in patients compared to controls. In addition, elevated levels of diacylglycerols and decreased cholesteryl esters were observed in patients. Using a machine learning model, we identified a signature of 38 biomarkers for PDAC, composed of 21 proteins, 4 lipids, and 13 metabolites. CONCLUSIONS Overall, this study identified several proteins, metabolites and lipids involved in various pathways including cholesterol and lipid metabolism to be changing in patients. In addition, we discovered a multi-analyte signature that could be further tested for detection of PDAC.
Collapse
Affiliation(s)
| | - Partho Sen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Caroline W Grant
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Roman Zenka
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Marimuthu Sappani
- Department of Biostatistics, Christian Medical College, Vellore, Tamil Nadu, 632002, India
| | - Jeyaseelan Lakshmanan
- College of Medicine, Mohammad Bin Rashid University of Medicine and Health Sciences, Dubai, 505055, UAE
| | - Arjun P Athreya
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Richard K Kandasamy
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
- Manipal Academy of Higher Education, Manipal, Karnataka, 5761904, India
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
- Manipal Academy of Higher Education, Manipal, Karnataka, 5761904, India.
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
11
|
Ji W, Xiong Y, Yang W, Shao Z, Guo X, Jin G, Su J, Zhou M. Transcriptomic profiling of blood platelets identifies a diagnostic signature for pancreatic cancer. Br J Cancer 2025:10.1038/s41416-025-02980-z. [PMID: 40133510 DOI: 10.1038/s41416-025-02980-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 02/26/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PaCa) is a deadly malignancy that is often diagnosed at an advanced stage, limiting treatment and reducing survival. There is an urgent need for convenient and accurate diagnostic markers for the early detection of PaCa. METHODS In this multicenter case-control study, we performed transcriptome analysis of 673 platelet samples from different in-house and public cohorts. RNA sequencing and RT-qPCR were used to discover and validate potential platelet biomarkers. A multi-gene signature was developed using binomial generalized linear model and independently validated in multicenter cohorts. RESULTS Two platelet RNAs, SCN1B and MAGOHB, consistently showed robust altered expression patterns between PaCa and healthy controls across cohorts, as confirmed by both RNA sequencing and RT-qPCR. The diagnostic two-RNA signature, PLA2Sig, demonstrated remarkable performance in detecting PaCa, with area under the receiver operating characteristic curve (AUC) values of 0.808, 0.900, 0.783, and 0.830 across multicenter cohorts. Furthermore, PLA2Sig effectively identified resectable stage I&II PaCa cases with an AUC of 0.812. Notably, PLA2Sig outperformed the traditional serum markers carcinoembryonic antigen and carbohydrate antigen 19-9 in distinguishing PaCa from healthy controls, and is complementary to established blood-based screening biomarkers. CONCLUSION These findings provide preliminary but promising evidence for the potential utility of platelet RNAs as an alternative non-invasive liquid biopsy tool for the early detection of PaCa.
Collapse
Affiliation(s)
- Weiping Ji
- Department of General Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yichun Xiong
- Department of General Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Wei Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230001, China
| | - Zhuo Shao
- Department of General Surgery, Shanghai Changhai Hospital of Navy Medical University, Shanghai, 200438, China
| | - Xiaoling Guo
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Gang Jin
- Department of General Surgery, Shanghai Changhai Hospital of Navy Medical University, Shanghai, 200438, China
| | - Jianzhong Su
- Department of General Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| | - Meng Zhou
- Department of General Surgery, School of Biomedical Engineering, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
12
|
Huynh HTLK, Lim HGM, Lee YCG, Phan TV, Vo TH, Chen CH, Wu ATH. In Silico Identification of ANKRD22 as a Theragnostic Target for Pancreatic Cancer and Fostamatinib's Therapeutic Potential. Int J Med Sci 2025; 22:1885-1904. [PMID: 40225855 PMCID: PMC11983316 DOI: 10.7150/ijms.105193] [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: 10/14/2024] [Accepted: 02/19/2025] [Indexed: 04/15/2025] Open
Abstract
Pancreatic cancer (PC) is one of the most tremendously malignant cancers with a poor prognosis, especially when it advances to metastasis. Besides, PC patients have encountered resistance to recent therapeutic approaches. In recent work, we effectively determined ANKRD22 by re-analyzing RNA-seq datasets from cell lines and human tissues deriving from PC. We demonstrated that ANKRD22 expression was remarkably high in the PC group compared to the normal group at both gene expression and protein levels. ANKRD22 resulted in a worse overall survival (OS) rate of PC patients (HR = 1.7, p = 0.0082). Intriguingly, ANKRD22 was statistically highly expressed in the mutated KRAS group relative to the wildtype group (p < 0.05). Similarly, compared to the wildtype TP53, in the mutated TP53, ANKRD22 also significantly expressed (p < 0.05); their concurrent expression, ANKRD22 and KRAS; ANKRD22 and TP53 exacerbated the survival outcome relative to the co-expression of low ANKRD22 and unaltered genes (p < 0.001; HR > 2.6). We explored the potential pathways and biological processes ANKRD22 might not only contribute to promoting PC, including cell-cycle regulation, E2F1 targets, and apoptosis but also foster the dissemination of PC by involve in invasion and migration processes. In the investigation of drugs that might target ANKRD22, we figured out fostamatinib. Molecular docking and molecular dynamic simulation (MDs) techniques provided extensive insights into the binding mode of ANKRD22 and fostamatinib. ANKRD22 exhibited strong binding affinity (ΔG = -7.0 kcal/mol in molecular docking and ∆Gbind = -38.66 ± 6.09 kcal/mol in MDs). Taken together, ANKRD22 could be a promising theragnostic target that might be inhibited by fostamatinib, thereby suppressing PC growth.
Collapse
Affiliation(s)
- Huong Thi Luu Kim Huynh
- International PhD Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Hendrick Gao-Min Lim
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Department of Medical Research, Tzu Chi Hospital Indonesia, Pantai Indah Kapuk, Greater Jakarta, Indonesia 14470
| | - Yuan-Chii Gladys Lee
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Thien-Vy Phan
- Department of Pharmacy, Nguyen Tat Thanh University, Ho Chi Minh City 700000, Vietnam
| | - Thanh-Hoa Vo
- University of Health Sciences, Vietnam National University Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Chien-Hsin Chen
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Colorectal Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Alexander T H Wu
- International PhD Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- The PhD Program of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan
- Taipei Heart Institute (THI), Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
13
|
Bayindir-Bilgic M, Duman E, Turgut D, Kadikoylu AN, Ekimci-Gurcan N, Ozbey U, Kuskucu A, Bayrak OF. Investigation of the synergistic effect of metformin and FX11 on PANC-1 cell lines. Biol Res 2025; 58:15. [PMID: 40091035 PMCID: PMC11912783 DOI: 10.1186/s40659-025-00592-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Pancreatic cancer is among the most aggressive and malignant tumors and is a leading cause of cancer-related mortality. It is characterized by its metabolic Warburg effect and glucose dependence. Aerobic glycolysis is a key feature of metabolic reprogramming in cancer cells. This study investigates the combined effect of metformin and FX11, hypothesizing that disrupting cancer cell energetics through complementary mechanisms may result in a synergistic therapeutic effect. The combination of metformin and FX11 affects the axis that regulates vital functions in cancer cells; thus, the uncontrolled growth of tumor cells, especially those that use a lactose-dependent energy pathway, can be controlled. Several in vitro experiments were conducted to evaluate this hypothesis. PANC-1 cell proliferation was assessed using an MTS assay, lactate levels were measured via an LDH assay, and apoptosis was determined using a flow cytometry-based PE-annexin V assay. The downstream effects of metformin and FX11 treatment were evaluated via western blot analysis. RESULTS The findings of this study revealed that metformin and FX11 significantly decreased the viability of PANC-1 cells when used in combination, and this effect was achieved by significantly affecting the energy mechanism of the cells through the AMPKα axis. Furthermore, the lactate levels in PANC1 cells co-treated with metformin and FX11 were significantly decreased, while the increased cellular stress led the cells to apoptosis. CONCLUSIONS Compared with metformin treatment alone, the combination treatment of metformin and FX11 stimulates cellular stress in pancreatic cancer and targets various energy processes that encourage cancer cells to undergo apoptosis. This study provides a novel therapeutic strategy for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Melike Bayindir-Bilgic
- Department of Medical Genetics, School of Medicine, Yeditepe University, Istanbul, Turkey
- Department of Genetics and Bioengineering, Yeditepe University, Acıbadem Mah. Liseyolu sok. No:8 Kat: 3, Kadıköy/Istanbul, 34718, Turkey
| | - Ezgi Duman
- Department of Medical Genetics, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Deniz Turgut
- Department of Medical Genetics, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Ayse Naz Kadikoylu
- Department of Medical Genetics, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Nur Ekimci-Gurcan
- Department of Medical Genetics, School of Medicine, Yeditepe University, Istanbul, Turkey
- Department of Genetics and Bioengineering, Yeditepe University, Acıbadem Mah. Liseyolu sok. No:8 Kat: 3, Kadıköy/Istanbul, 34718, Turkey
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Biruni University, Istanbul, Turkey
| | - Utku Ozbey
- Department of Medical Genetics, School of Medicine, Yeditepe University, Istanbul, Turkey
- Department of Genetics and Bioengineering, Yeditepe University, Acıbadem Mah. Liseyolu sok. No:8 Kat: 3, Kadıköy/Istanbul, 34718, Turkey
| | - Aysegul Kuskucu
- Department of Medical Genetics, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Omer F Bayrak
- Department of Medical Genetics, School of Medicine, Yeditepe University, Istanbul, Turkey.
- Department of Genetics and Bioengineering, Yeditepe University, Acıbadem Mah. Liseyolu sok. No:8 Kat: 3, Kadıköy/Istanbul, 34718, Turkey.
| |
Collapse
|
14
|
Pal RS, Jawaid T, Rahman MA, Verma R, Patra PK, Vijaypal SV, Pal Y, Upadhyay R. Metformin's anticancer odyssey: Revealing multifaceted mechanisms across diverse neoplastic terrains- a critical review. Biochimie 2025; 233:109-121. [PMID: 40058683 DOI: 10.1016/j.biochi.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/10/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Metformin, initially prescribed as an oral hypoglycemic medication for type 2 diabetes, has recently gained attention for its potential anticancer effects. Its history dates to 1918, when guanidine, a component of the traditional European herb Galega officinalis, was found to reduce glycemia. This review precisely examines the mechanisms underlying Metformin's anticancer effects across various neoplastic conditions. This investigation explores the complex interactions between metformin and major signaling pathways associated with carcinogenesis, including AMP-activated protein kinase (AMPK), mTOR, and insulin-like growth factor (IGF) pathways. The review emphasizes Metformin's diverse effects on angiogenesis, inflammation, apoptosis, and cellular metabolism in cancer cells. Additionally, new data on metformin's capacity to alter the tumor microenvironment and enhance immune surveillance systems against cancer are examined. The review underscores Metformin's potential for repurposing in oncology, emphasizing its clinical relevance as an adjuvant therapy for various cancers. The review provides insightful information about the complex anticancer mechanisms of metformin by combining data from preclinical and clinical studies. These findings not only broaden our knowledge of the effects of metformin but also open new avenues for oncology research and treatment developments.
Collapse
Affiliation(s)
- Rashmi Saxena Pal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Talha Jawaid
- Department of Pharmacology, College of Medicine, Imam Muhammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - M A Rahman
- Teegala Krishna Reddy College of Pharmacy, Hyderabad, Telangana, India
| | - Rakesh Verma
- Department of Pharmacology, Institute of Medical Science, BHU, Varanasi, Uttar Pradesh, India
| | - Pratap Kumar Patra
- School of Pharmacy & Life Sciences, Centurion University of Technology & Management, Bhubaneswar, Odisha, India
| | | | - Yogendra Pal
- School of Pharmaceutical Science, RIMT University, Mandi Gobindgarh, Punjab, India
| | - Rohit Upadhyay
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
15
|
Yang H, Chou W, Nguyen P, Nguyen NTH, Phuong NT, Wang C, Hsu JC, Lin M, Huang C. The protective role of anti-parkinsonian drugs in pancreatic cancer risk: A comprehensive case-control study in Taiwan. Cancer Sci 2025; 116:783-791. [PMID: 39629516 PMCID: PMC11875767 DOI: 10.1111/cas.16422] [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: 09/09/2024] [Revised: 11/01/2024] [Accepted: 11/20/2024] [Indexed: 03/05/2025] Open
Abstract
Pancreatic cancer is among the deadliest cancers, with a grim prognosis despite advances in treatment. We conducted a population-based case-control study from Taiwan, linking Health and Welfare Data Science Center data to the Taiwan Cancer Registry, which offers a promising strategy for its treatment through drug repurposing. The study aims to identify the association of anti-parkinsonian drugs with pancreatic cancer risk across different age groups. The analysis encompassed 18,921 pancreatic cancer cases and 75,684 matched controls, employing conditional logistic regression to assess the impact of anti-parkinsonian drugs on the risk of pancreatic cancer. Key findings revealed a statistically significant association of the administration with specific anti-parkinsonian medications, including anticholinergic agents, tertiary amines, dopa derivatives, and dopamine receptor agonists, with a reduction in pancreatic cancer risk. These associations were represented as adjusted odds ratios (aORs), ranging from 0.620 (95% CI 0.470-0.810) to 0.764 (95% CI 0.655-0.891). Further, age-stratified analysis revealed variations in efficacy across different age groups. Anticholinergic agents and tertiary amines exhibited greater effectiveness in the 40-64-year age group (aOR, 0.653; 95% CI, 0.489-0.872), whereas dopa derivatives and dopamine receptor agonists were particularly efficacious in the cohort aged ≥65 years (aOR, 0.728; 95% CI, 0.624-0.850 and aOR, 0.665; 95% CI, 0.494-0.894, respectively). Notably, specific drugs such as trihexyphenidyl, levodopa/dopa decarboxylase inhibitor (DDCI), and pramipexole demonstrated a significant decrease in cancer risk, especially in the elderly population. These preliminary findings can contribute to the possible therapeutic role of anti-parkinsonian drugs in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Hsuan‐Chia Yang
- Graduate Institute of Biomedical Informatics, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- International Center for Health Information Technology (ICHIT), College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- Clinical Big Data Research Center, Taipei Medical University HospitalTaipei Medical UniversityTaipeiTaiwan
- Research Center of Big Data and Meta‐Analysis, Wan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Wen‐Chi Chou
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou and College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Phung‐Anh Nguyen
- Clinical Big Data Research Center, Taipei Medical University HospitalTaipei Medical UniversityTaipeiTaiwan
- Graduate Institute of Data Science, College of ManagementTaipei Medical UniversityTaipeiTaiwan
- Clinical Data Center, Office of Data ScienceTaipei Medical UniversityTaipeiTaiwan
- Research Center of Health Care Industry Data Science, College of ManagementTaipei Medical UniversityTaipeiTaiwan
| | - Nhi Thi Hong Nguyen
- School of Nutrition and Health SciencesTaipei Medical UniversityTaipeiTaiwan
- Health Personnel Training InstituteUniversity of Medicine and Pharmacy, Hue UniversityHueVietnam
| | - Nguyen Thi Phuong
- Graduate Institute of Biomedical Informatics, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
| | - Ching‐Huan Wang
- Biomedical Informatics and Data Science (BIDS) Section, School of MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jason C. Hsu
- Clinical Big Data Research Center, Taipei Medical University HospitalTaipei Medical UniversityTaipeiTaiwan
- Clinical Data Center, Office of Data ScienceTaipei Medical UniversityTaipeiTaiwan
- Research Center of Health Care Industry Data Science, College of ManagementTaipei Medical UniversityTaipeiTaiwan
- International Ph.D. Program in Biotech and Healthcare Management, College of ManagementTaipei Medical UniversityTaipeiTaiwan
| | - Ming‐Chin Lin
- Graduate Institute of Biomedical Informatics, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- Department of Neurosurgery, Wang‐Fang HospitalTaipei Medical UniversityTaipeiTaiwan
- Taipei Neuroscience InstituteTaipei Medical UniversityTaipeiTaiwan
| | - Chih‐Wei Huang
- Graduate Institute of Biomedical Informatics, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- International Center for Health Information Technology (ICHIT), College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- Clinical Big Data Research Center, Taipei Medical University HospitalTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
16
|
Zhou Y, Lee HH, Tang Y, Yu X, Yang Q, Kim ME, Remedios LW, Bao S, Spraggins JM, Huo Y, Landman BA. Multi-contrast computed tomography atlas of healthy pancreas with dense displacement sampling registration. J Med Imaging (Bellingham) 2025; 12:024006. [PMID: 40255249 PMCID: PMC12005954 DOI: 10.1117/1.jmi.12.2.024006] [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: 08/19/2024] [Revised: 03/26/2025] [Accepted: 03/26/2025] [Indexed: 04/22/2025] Open
Abstract
Purpose Diverse population demographics can lead to substantial variation in the human anatomy. Therefore, standard anatomical atlases are needed for interpreting organ-specific analyses. Among abdominal organs, the pancreas exhibits notable variability in volumetric morphology, shape, and appearance, complicating the generalization of population-wide features. Understanding the common features of a healthy pancreas is crucial for identifying biomarkers and diagnosing pancreatic diseases. Approach We propose a high-resolution CT atlas framework optimized for the healthy pancreas. We introduce a deep-learning-based preprocessing technique to extract abdominal ROIs and leverage a hierarchical registration pipeline to align pancreatic anatomy across populations. Briefly, DEEDS affine and non-rigid registration techniques are employed to transfer patient abdominal volumes to a fixed high-resolution atlas template. To generate and evaluate the pancreas atlas, multi-phase contrast CT scans of 443 subjects (aged 15 to 50 years, with no reported history of pancreatic disease) were processed. Results The two-stage DEEDS affine and non-rigid registration outperforms other state-of-the-art tools, achieving the highest scores for pancreas label transfer across all phases (non-contrast: 0.497, arterial: 0.505, portal venous: 0.494, delayed: 0.497). External evaluation with 100 portal venous scans and 13 labeled abdominal organs shows a mean Dice score of 0.504. The low variance between the pancreases of registered subjects and the obtained pancreas atlas further illustrates the generalizability of the proposed method. Conclusion We introduce a high-resolution pancreas atlas framework to generalize healthy biomarkers across populations with multi-contrast abdominal CT. The atlases and the associated pancreas organ labels are publicly available through the Human Biomolecular Atlas Program (HuBMAP).
Collapse
Affiliation(s)
- Yinchi Zhou
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
| | - Ho Hin Lee
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
| | | | - Xin Yu
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
| | - Qi Yang
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
| | - Michael E. Kim
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
| | - Lucas W. Remedios
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
| | - Shunxing Bao
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
| | - Jeffrey M. Spraggins
- Vanderbilt University, Department of Cell and Developmental Biology, Nashville, Tennessee, United States
- Vanderbilt University, Department of Biochemistry, Nashville, Tennessee, United States
- Vanderbilt University, Department of Chemistry, Nashville, Tennessee, United States
- Vanderbilt University Medical Center, Department of Pathology, Microbiology, and Immunology, Nashville, Tennessee, United States
- Vanderbilt University Medical Center, Department of Radiology, Nashville, Tennessee, United States
| | - Yuankai Huo
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
- Vanderbilt University, Department of Electrical and Computer Engineering, Nashville, Tennessee, United States
| | - Bennett A. Landman
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
- Vanderbilt University Medical Center, Department of Radiology, Nashville, Tennessee, United States
- Vanderbilt University, Department of Electrical and Computer Engineering, Nashville, Tennessee, United States
| |
Collapse
|
17
|
Kikuyama M, Nakahodo J, Chiba K, Honda G. Focal pancreatic parenchymal atrophy could be a precursor of pancreatic cancer. Pancreatology 2025; 25:241-249. [PMID: 39894733 DOI: 10.1016/j.pan.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 12/10/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND/OBJECTIVES We previously reported that focal pancreatic parenchymal atrophy (FPPA) indicates high-grade pancreatic intraepithelial neoplasia (HG-PanIN) or carcinoma in situ (CIS). Because HG-PanIN progresses into pancreatic ductal adenocarcinoma (PDAC), the relationship between FPPA and PDAC should be investigated. METHODS We included 54 patients with PDAC, whose previous computed tomography or magnetic resonance imaging were reviewed. The existence, positional relationship between FPPA and PDAC, and time between FPPA recognition and PDAC diagnosis were all examined. Of the 54 patients, 28 underwent surgery. The remaining 26 patients were histopathologically diagnosed with PDAC using endoscopic ultrasonography-guided fine needle aspiration. RESULTS Among the 54 patients included, 49 (83.3 %) had FPPA. The pancreatic head and body were the common sites of FPPA. In all patients with FPPA, PDAC developed near the FPPA, with an average distance of 7.93 mm between the edge of the FPPA and the center of the PDAC. The interval between FPPA recognition and PDAC diagnosis was 35.33 months, which was significantly shorter in the surgical group. CONCLUSIONS FPPA could be a precursor of PDAC and suggest the area at risk of PDAC.
Collapse
Affiliation(s)
- Masataka Kikuyama
- Department of Gastroenterology, Tokyo Women's Medical University Hospital, Tokyo, Japan.
| | - Jun Nakahodo
- Department of Gastroenterology, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | - Kazuro Chiba
- Department of Gastroenterology, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | - Goro Honda
- Department of Surgery, Tokyo Women's Medical University Hospital, Tokyo, Japan
| |
Collapse
|
18
|
Luo R, Li S, Yang C, Tang B, Li L, Luo C. Curcumin Inhibits the Development of Pancreatic Cancer by Targeting the circ_0079440/miR-522-3p/EIF4A1 Pathway. Cell Biochem Biophys 2025; 83:377-390. [PMID: 39102088 DOI: 10.1007/s12013-024-01466-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 08/06/2024]
Abstract
Pancreatic cancer (PC) is a common gastrointestinal cancer with high invasiveness and high mortality. Curcumin is a natural polyphenol with anti-tumor activity against different cancers, including PC. Curcumin has been verified to mediate the expression of circular RNAs (circRNAs) to inhibit tumor development. This study aimed to explore the function and regulatory mechanism of curcumin on circ_0079440 in PC. PC cells were treated with different concentrations of curcumin (0, 5, 10 or 15 μM) for 24 h. Gene expression in PC cells and tissues was detected using RT-qPCR. Cell malignant phenotypes were determined by functional assays. The levels of EMT-related proteins were tested using western blot. RNA interaction was determined using RNA pulldown assay, luciferase reporter assay and RIP assay. The results showed that curcumin suppressed cell proliferative, migratory, and invasive capabilities, and weakened epithelial-mesenchymal transition (EMT) in a concentration-dependent way. Circ_0079440 was expressed at a high level in PC and its level was reduced via curcumin administration in PC cells. Rescue assays showed that circ_0079440 overexpression reversed the suppressive effects of curcumin on PC cell malignant phenotypes. Furthermore, in the xenograft mouse models, curcumin treatment inhibited tumor growth and metastasis, and circ_0079440 upregulation reversed the function of curcumin. Additionally, circ_0079440 was revealed to bind to miR-522-3p to upregulate eukaryotic initiation factor 4A1 (EIF4A1) expression in PC cells. EIF4A1 expression was also downregulated by curcumin, and EIF4A1 overexpression abolished the suppressive functions of curcumin. Moreover, EIF4A overexpression or miR-522-3p inhibition counteracted the anti-tumor effects of circ_0079440 depletion on PC development. To sum up, curcumin suppresses PC development by targeting the circ_0079440/miR-522-3p/EIF4A1 pathway, which might provide novel therapeutic targets for treatment of PC.
Collapse
Affiliation(s)
- Ruiying Luo
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Shuang Li
- Department of respiratory medicine, The Third People's Hospital of Gansu Province, Lanzhou, 730000, Gansu, China
| | - Chi Yang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Baoyuan Tang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Long Li
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Changjiang Luo
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
19
|
Ul Hassan MH, Shahbaz M, Imran M, Momal U, Naeem H, Mujtaba A, Hussain M, Anwar MJ, Alsagaby SA, Al Abdulmonem W, Yehuala TF, Abdelgawad MA, El‐Ghorab AH, Selim S, Mostafa EM. Isoflavones: Promising Natural Agent for Cancer Prevention and Treatment. Food Sci Nutr 2025; 13:e70091. [PMID: 40078339 PMCID: PMC11896816 DOI: 10.1002/fsn3.70091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Isoflavones are currently being investigated by researchers in order to demonstrate their ability to prevent the proliferation of cancer cells. The current review aimed to demonstrate the potential of isoflavones to eliminate cancerous cells in the stomach, liver, lung, breast, and prostate, as their anticancer properties are due to the ability to block the signaling pathways of the extracellular signal-controlled kinase (MAPK/ERK) and proteasome (PI3K/AKT/mTOR). Isoflavones can inhibit the cell division of various cancer cells. Isoflavones can block the androgen receptor (AR), a protein that is required for the growth and dissemination of prostate cancer. It initiates the caspase cascade and obstructs the production of new proteins to eliminate lung cancer cells. These inhibit colon cancer cells by entering their G2/M cell cycle phase and inducing apoptosis. These are also known to inhibit the production of cyclin-dependent kinase 2 and cyclin B1, two proteins that are related to an enhanced risk of colon cancer. These suppress the breakdown of cyclin B1 and CDK2 to stop the development of cancer. Preclinical evidence consistently supports the efficacy of isoflavones in suppressing tumor growth; however, human clinical trials show variability due to differences in bioavailability, metabolism, and dosage. Despite their promise as alternative or adjunctive cancer therapies, limitations such as low solubility, interindividual metabolic variations, and inconsistent clinical outcomes necessitate further large-scale, controlled trials. Future research should focus on improving bioavailability and exploring synergistic effects with conventional therapies.
Collapse
Affiliation(s)
- Muhammad Hammad Ul Hassan
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of AgricultureMultanPakistan
| | - Muhammad Shahbaz
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of AgricultureMultanPakistan
| | - Muhammad Imran
- Department of Food Science and TechnologyUniversity of NarowalNarowalPakistan
| | - Ushna Momal
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of AgricultureMultanPakistan
| | - Hammad Naeem
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of AgricultureMultanPakistan
- Post Harvest Research CentreAyub Agricultural Research InstituteFaisalabadPakistan
| | - Ahmed Mujtaba
- Department of Food Science and Technology, Faculty of Engineering Sciences and TechnologyHamdard University Islamabad CampusIslamabadPakistan
| | - Muzzamal Hussain
- Department of Food ScienceGovernment College University FaisalabadFaisalabadPakistan
| | - Muhammad Junaid Anwar
- Department of Food Science and Technology, Faculty of Food Science and NutritionBahauddin Zakariya UniversityMultanPakistan
| | - Suliman A. Alsagaby
- Department of Medical Laboratory Sciences, College of Applied Medical SciencesMajmaah UniversityAl‐MajmaahSaudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of MedicineQassim UniversityBuraidahSaudi Arabia
| | - Tadesse Fenta Yehuala
- Faculty of Chemical and Food Engineering, Bahir Dar Institute of TechnologyBahir Dar UniversityBahir Dar CityEthiopia
| | - Mohamed A. Abdelgawad
- Department of Pharmaceutical Chemistry, College of PharmacyJouf UniversitySakakaAljoufSaudi Arabia
| | - Ahmed H. El‐Ghorab
- Department of Chemistry, College of ScienceJouf UniversitySakakaSaudi Arabia
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesJouf UniversitySakakaSaudi Arabia
| | - Ehab M. Mostafa
- Department of Pharmacognosy, College of PharmacyJouf UniversitySakakaSaudi Arabia
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys)Al‐Azhar UniversityCairoEgypt
| |
Collapse
|
20
|
Darbandi M, Khorrami Z, Karamoozian A, Aboubakri O, Miryan M, Rezakhani L, Shadmani FK. A comparison of the burden of cancers between 1990 and 2019 in Iran: A national and subnational study. PLoS One 2025; 20:e0309699. [PMID: 39999060 PMCID: PMC11856284 DOI: 10.1371/journal.pone.0309699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 08/17/2024] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Cancer is a rapidly increasing global problem, and one of the leading causes of burden and mortality. This study aims to compare the burden of cancer in Iran between the year 1990 and 2019. METHODS We used Global Burden of Disease data on cancer from 1990 to 2019 by province, year, age group, and sex. We then estimated the trend of age standardized mortality and Disability-Adjusted Life Years (DALYs) of the cancers by sex. Age pattern and geographical variation in the ranking of cancers were assessed at national and sub-national levels from 1990 to 2019. RESULTS The mortality rate decreased from 102 (95% UI: 91, 111) to 96 (95% UI: 88, 103) per 100000 population. Additionally, the DALYs rates decreased from 2619 (95% UI: 2357, 2852) to 2321 (95% UI: 2116, 2497) per 100000 between 1990 and 2019. Both of the mortality and DALYs rate from cancers increased with age. These indicators were significantly higher in men than in women across all age groups. Consequently, the mortality rate and DALYs per 100,000 of cancers were higher in the northwest and northeast of Iran. Notably, stomach cancer was identified as the leading cause of cancer mortality in 23 provinces of Iran in 2019. The highest percentage change of DALYs per 100,000 rate between 1990 and 2019 was observed for malignant skin melanoma, stomach cancer, and cervical cancers with rate of -41.1, -40.1, and -38.4, respectively. CONCLUSION Overall, the mortality and DALYs per 100,000 rates of all cancers for both sexes in Iran have decreased between 1990 and 2019. However, there is an increasing trend in types of cancers, such as pancreatic, ovarian, and breast cancers.
Collapse
Affiliation(s)
- Mitra Darbandi
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Khorrami
- Ophthalmic Epidemiology Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Karamoozian
- Modeling in Health Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
- Department of Biostatistics and Epidemiology, Kerman University of Medical Sciences, Kerman, Iran
| | - Omid Aboubakri
- Environmental Health Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mahsa Miryan
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Khosravi Shadmani
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
21
|
Mahdian SMA, Mahmoudi-Aznaveh A, Mousavi SM, Larijani B, Azizi Z, Javar HA. Plasma treatment can efficiently increase the attachment of pancreatic circulatory tumor cells to the surface. Discov Oncol 2025; 16:222. [PMID: 39982607 PMCID: PMC11845332 DOI: 10.1007/s12672-025-01988-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Pancreatic cancer ranks as the fourth most common cause of cancer-related fatalities globally, with a notably low 5-year relative survival rate. We need to immediately develop fast, dependable, and noninvasive diagnostic techniques that can accurately identify pancreatic cancer at an early stage. The research project created a straightforward but effective method for detecting and increasing the amount of tumor cells that could bind to polystyrene (PS) well plates. To significantly improve the adhesion of the pancreatic cancer cell line PANC-1 on PS well plates, a 5-min exposure to high-power oxygen plasma was implemented. This treatment caused a significant increase in surface energy and roughness. Surface characterization was assessed by utilizing an atomic force microscope and X-ray photoelectron spectroscopy. Water contact angle measurement is used to assess the level of wettability present on the treated surface. To determine how well the circulatory tumor cells (CTCs) model adheres to a plasma-treated surface (PTS), appropriate amounts of mCherry-labeled PANC-1 cells are mixed into a sample of blood cells to mimic clinical conditions. After applying plasma treatment, the experiment achieved a 96% success rate in binding at 2 h, specifically for the PANC-1 cell type. Moreover, the platform demonstrated a considerable ability to attach to cancerous cells compared to non-cancerous cells found in blood. To summarize, this study has shown that non-thermal plasma treatment could be a novel and efficient method for the better adhesion of pancreatic cancer cells, with the benefits of being cost-effective and quick. It is necessary for additional research to be conducted to confirm the clinical efficacy of the method.
Collapse
Affiliation(s)
- Seyed Mohammad Amin Mahdian
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Azam Mahmoudi-Aznaveh
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mojtaba Mousavi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Azizi
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hamid Akbari Javar
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Di Marco F, Cufaro MC, Damiani V, Dufrusine B, Pizzinato E, Di Ferdinando F, Sala G, Lattanzio R, Dainese E, Federici L, Ponsaerts P, De Laurenzi V, Cicalini I, Pieragostino D. Proteomic meta-analysis unveils new frontiers for biomarkers research in pancreatic carcinoma. Oncogenesis 2025; 14:3. [PMID: 39956821 PMCID: PMC11830788 DOI: 10.1038/s41389-025-00547-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/20/2024] [Accepted: 02/06/2025] [Indexed: 02/18/2025] Open
Abstract
Pancreatic carcinoma (PC) is the sixth leading cause of cancer death in both sexes in 2022, responsible for almost 5% of all cancer deaths worldwide; it is characterized by a poor prognosis since most patients present with an unresectable and metastatic tumor. To date, the decreasing trend in mortality rates related to the most common cancers has contributed to making pancreatic cancer a serious public health problem. In the last few years, scientific research has led to many advances in diagnostic approaches, perioperative management, radiotherapy techniques, and systemic therapies for advanced disease, but only with modest incremental progress in PC patient outcomes. Most of the causes of this high mortality are, unfortunately, late diagnosis and an important therapeutic resistance; for this reason, the most recent high-throughput proteomics technologies focus on the identification of novel biomarkers and molecular profiling to generate new insights in the study of PC, to improve diagnosis and prognosis and to monitor the therapies progress. In this work, we present and discuss the integration of results from different revised studies on protein biomarkers in a global proteomic meta-analysis to understand which path to pursue scientific research. In particular, cancer signaling, inflammatory response, and cell migration and signaling have emerged as the main pathways described in PC, as well as scavenging of free radicals and metabolic alteration concurrently highlighted new research insights on this disease. Interestingly, from the study of upstream regulators, some were found to be shared by collecting data relating to both biological fluid and tissue biomarkers, side by side: specifically, TNF, LPS, p38-MAPK, AGT, miR-323-5p, and miR-34a-5p. By integrating many biological components with their interactions and environmental relationships, it's possible to achieve an in-depth description of the pathological condition in PC and define correlations between concomitant symptoms and tumor genesis and progression. In conclusion, our work may represent a strategy to combine the results from different studies on various biological samples in a more comprehensive way.
Collapse
Affiliation(s)
- Federica Di Marco
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Maria Concetta Cufaro
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Verena Damiani
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Beatrice Dufrusine
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Erika Pizzinato
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Telematic University of "Leonardo Da Vinci", Torrevecchia Teatina, Chieti, Italy
| | - Fabio Di Ferdinando
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Gianluca Sala
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Rossano Lattanzio
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Enrico Dainese
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Luca Federici
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerpen, Belgium
| | - Vincenzo De Laurenzi
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Ilaria Cicalini
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy.
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy.
| | - Damiana Pieragostino
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
23
|
Kobayashi K, Kishi Y, Tsunenari T, Yonamine N, Takihata Y, Nakazawa A, Takao M, Einama T, Tsujimoto H, Ueno H. Risk of hepatic steatosis with the preoperative treatment of pancreatic cancer and the short-term postoperative outcomes. Surg Today 2025; 55:211-221. [PMID: 38980333 DOI: 10.1007/s00595-024-02895-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: 03/16/2024] [Accepted: 06/19/2024] [Indexed: 07/10/2024]
Abstract
PURPOSE We investigated whether the preoperative treatment of patients with pancreatic cancer is a risk factor for hepatic steatosis (HS), and whether preoperative HS affects the short-term postoperative outcomes. METHODS Patients who underwent radical surgery for pancreatic cancer between 2010 and 2023 were enrolled. The patients' medical records were reviewed. Albumin and carbohydrate antigen 19-9 were measured before and after chemotherapy in the patients who received preoperative chemotherapy. A logistic regression univariate analysis was performed to analyze the factors associated with new-onset HS. RESULTS A total of 230 patients who underwent surgery were included. HS was observed on the date of surgery in 11 (10%) and two (2%) patients with and without preoperative chemotherapy, respectively. Female sex, initially borderline resectable or unresectable disease, history of cholangitis, presence of PEI, long-term (≥ 3 months) biliary drainage, preoperative chemotherapy, and serum albumin ≥ 3.9 mg/dl before chemotherapy were identified as risk factors for HS. The incidence of postoperative morbidity did not differ between the patients with and without preoperative steatosis. CONCLUSIONS Preoperative chemotherapy, a history of cholangitis, the presence of PEI, and ≥ 3 months' duration of biliary drainage were risk factors for the development of HS before surgery for pancreatic cancer. However, preoperative HS did not affect the short-term postoperative outcomes.
Collapse
Affiliation(s)
- Kazuki Kobayashi
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Yoji Kishi
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| | - Takazumi Tsunenari
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Naoto Yonamine
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Yasuhiro Takihata
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Akiko Nakazawa
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Mikiya Takao
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Takahiro Einama
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Hironori Tsujimoto
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| |
Collapse
|
24
|
Saito K, Nakai Y, Sasaki T, Takeda T, Ueno M, Tezuka S, Isayama H, Tomishima K, Kojima Y, Yamamoto N, Ito Y, Oyama H, Toda N, Takagi K, Matsubara S, Mohri D, Sato T, Fujishiro M. Impact of Statin Use on Survival in Patients With Unresectable Pancreatic cancer Receiving Gemcitabine Plus Nab-Paclitaxel: A Multicenter Retrospective Study. Pancreas 2025; 54:e107-e113. [PMID: 39928888 DOI: 10.1097/mpa.0000000000002411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
PURPOSE The aim of this multicenter retrospective study was to evaluate the impact of statin use on clinical outcomes in patients with unresectable pancreatic cancer (PC) receiving gemcitabine plus nab-paclitaxel (GnP) in a large Japanese cohort. MATERIALS AND METHODS We retrospectively reviewed the medical records including data on the use of concomitant medications in patients with unresectable PC receiving GnP between January 2015 and January 2019 at 10 hospitals. Prognostic factors for progression-free survival (PFS) and overall survival (OS) were evaluated. RESULTS A total of 1682 patients were included in the analysis; of which 322 patients (19%) received statins and 1360 (81%) did not receive statin. The median PFS and OS were 7.5 versus 7.3 months (P = 0.87) and 15.1 versus 14.4 months (P = 0.48) in cases with and without statin use. The use of statin was not associated with PFS (hazard ratio, 1.01; 95% confidence interval, 0.85-1.18, P = 0.93) or OS (hazard ratio, 1.05; 95% confidence interval, 0.91-1.21, P = 0.47) in the multivariable analyses. PFS and OS did not significantly differ by liposolubility of statins, either. CONCLUSIONS Stain use was not associated with PFS or OS in patients with unresectable PC receiving GnP.
Collapse
Affiliation(s)
| | | | - Takashi Sasaki
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tsuyoshi Takeda
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Shun Tezuka
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Hiroyuki Isayama
- Department of Gastroenterology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Ko Tomishima
- Department of Gastroenterology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yasushi Kojima
- Department of Gastroenterology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Natsuyo Yamamoto
- Department of Gastroenterology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yukiko Ito
- Department of Gastroenterology, Japanese Red Cross Medical Center, Tokyo, Japan
| | | | - Nobuo Toda
- Department of Gastroenterology, Mitsui Memorial Hospital, Tokyo, Japan
| | - Kaoru Takagi
- Department of Gastroenterology, Mitsui Memorial Hospital, Tokyo, Japan
| | - Saburo Matsubara
- Department of Gastroenterology and Hepatology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Dai Mohri
- Department of Gastroenterology, JR Tokyo general hospital, Tokyo, Japan
| | | | - Mitsuhiro Fujishiro
- From the Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
25
|
Sayeed MS, Ekambaram G, Rajput JPS, Parmar P, Saikhedkar S, Saiyad SS, Saiyad T. The Impact of Biliary Drainage on Quality of Life in Unresectable Biliary and Pancreatic Cancer. Cureus 2025; 17:e78986. [PMID: 40099061 PMCID: PMC11911705 DOI: 10.7759/cureus.78986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Obstructive jaundice, a frequent complication of biliary tract and pancreatic malignancies, severely impacts patients' quality of life (QoL). This condition is prevalent in unresectable cancers, often causing symptoms such as pruritus, anorexia, nausea, vomiting, and cholangitis, which hinder daily activities. Given the global burden of these cancers, the primary aim of the study was to evaluate the impact of biliary drainage on QoL and pruritus in patients with unresectable biliary and pancreatic malignancies. Secondary objectives include assessing changes in individual QoL domains and identifying factors associated with QoL outcomes. Materials and methods This prospective observational study was conducted at Meenakshi Mission Hospital and Research Centre, Madurai, from July 2020 to November 2021. The study included 82 patients with inoperable malignant biliary obstruction presenting with obstructive jaundice. Participants underwent either endoscopic retrograde cholangiopancreatography (ERCP; n=47, 57.3%) or percutaneous transhepatic biliary drainage (PTBD; n=35, 42.7%), depending on clinical suitability. QoL and pruritus were assessed at baseline, 1 week, and 4 weeks using validated tools (Functional assessment of cancer therapy-General (FACT-G), Functional assessment of cancer therapy-Hepatobiliary subscale (FACT-HS), and visual analog scale for pruritus (VASP)). Statistical analysis was performed using SPSS v26.0, with significance set at p < 0.05. Results and discussion The study involved 82 patients diagnosed with inoperable malignant biliary obstruction and presenting with obstructive jaundice. The mean age of the participants was 63.7 ± 10.5 years, with 58 (70.7%) of them being male. Type 2 diabetes (63.4%) and hypertension (52.4%) were prevalent comorbidities. Most patients had moderate functional impairment (64.6%). Biliary drainage led to a significant reduction in bilirubin levels, decreasing from 13 mg/dL to 1.3 mg/dL by the fourth week (p < 0.001) but reducing social well-being (p < 0.001). FACT-G, FACT-HS, and hepatobiliary scores declined despite pruritus relief (p = 0.001). While emotional well-being (EWB) improved initially, its effect was not sustained at 4 weeks (p = 0.084), indicating persistent psychological distress. Despite symptomatic relief, overall QoL declined, emphasizing the need for a multidisciplinary approach incorporating palliative care, nutritional support, and psychological counseling to improve long-term patient outcomes. Conclusion The study demonstrates that while biliary drainage significantly improves physical well-being, as evidenced by the reduction in bilirubin levels, there is a decline in the other aspects of QoL, highlighting the complexity of symptom management in patients with malignant biliary obstruction. These findings highlight the need for a comprehensive approach that addresses multiple aspects of well-being in this patient population.
Collapse
Affiliation(s)
| | - Gnanadesigan Ekambaram
- Department of Physiology, Nootan Medical College and Research Centre, Sankalchand Patel University, Visnagar, IND
| | | | - Pooja Parmar
- Department of Anatomy, J and D Institute of Nursing, Surat, IND
| | - Swapnil Saikhedkar
- Department of Physiology, Pacific Medical College and Hospital, Udaipur, IND
| | - Sajidali S Saiyad
- Department of Physiology, Pacific Medical College and Hospital, Udaipur, IND
| | - Tehsin Saiyad
- Department of Microbiology, Veer Narmad University, Surat, IND
| |
Collapse
|
26
|
Tan Z, Meng Y, Wu Y, Zhen J, He H, Pu Y, Zhang J, Dong W. The burden and temporal trend of early onset pancreatic cancer based on the GBD 2021. NPJ Precis Oncol 2025; 9:32. [PMID: 39880919 PMCID: PMC11779834 DOI: 10.1038/s41698-025-00820-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
In the context of the global increase in early-onset tumours, investigating the global disease burden caused by early-onset pancreatic cancer (EOPC) is imperative. Data on the burden of EOPC were obtained from the Global Burden of Disease Study 2021. A joinpoint regression model was used to analyse the temporal trend of the EOPC burden, and an age‒period‒cohort (APC) model was used to analyse the influence of age, period, and birth cohort on burden trends. Globally, the number of EOPC cases increased from 24,480 to 42,254, and the number of deaths increased from 17,193 to 26,996 between 1990 and 2021. The results of the APC model showed that the burden of EOPC increases with increasing age, whereas the variations in period and cohort effects exhibited a complex pattern across different sociodemographic index regions. Consequently, the disease burden of EOPC is increasing worldwide, highlighting the need for effective interventions.
Collapse
Affiliation(s)
- Zongbiao Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Yang Meng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yanrui Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Junhai Zhen
- Department of General Practice, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, China
| | - Haodong He
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Yu Pu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Jixiang Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
| |
Collapse
|
27
|
Chen Y, Ye X, Hu M, Hu Y, Ding J. Long non-coding RNAs in pancreatic cancer. Clin Chim Acta 2025; 566:120040. [PMID: 39536894 DOI: 10.1016/j.cca.2024.120040] [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: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
This article reviews the recent advances in pathogenesis, diagnosis and treatment of pancreatic cancer, as well as the relationship between long non-coding RNA (lncRNA) in disease progression. Unfortunately, pancreatic cancer has no early symptoms and quickly invades surrounding tissue and organs, making it one of the deadliest. Accordingly, we urgently need to identify high-risk individuals with precancerous lesions through screening methods to identify early disease, provide better prevention strategies and improve overall survival. LncRNAs have a variety of biological functions in both physiologic and pathophysiologic states including tumor growth, differentiation and proliferation. Herein we review the biological functions, expression patterns, clinical significance and targeted therapy potential of lncRNAs to provide new approaches for diagnosis and treatment in pancreatic cancer.
Collapse
Affiliation(s)
- Yuan Chen
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China
| | - Xiaohua Ye
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China
| | - Minli Hu
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China
| | - Yibing Hu
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China
| | - Jin Ding
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, Zhejiang, China.
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Pian LL, Song MH, Wang TF, Qi L, Peng TL, Xie KP. Identification and analysis of pancreatic intraepithelial neoplasia: opportunities and challenges. Front Endocrinol (Lausanne) 2025; 15:1401829. [PMID: 39839479 PMCID: PMC11746065 DOI: 10.3389/fendo.2024.1401829] [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: 03/16/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Pancreatic intraepithelial neoplasia (PanIN) is the most common precursor lesion of pancreatic ductal adenocarcinoma (PDAC), which has poor prognosis with a short median overall survival of 6-12 months and a low 5-year survival rate of approximately 3%. It is crucial to remove PanIN lesions to prevent the development of invasive PDAC, as PDAC spreads rapidly outside the pancreas. This review aims to provide the latest knowledge on PanIN risk, pathology, cellular origin, genetic susceptibility, and diagnosis, while identifying research gaps that require further investigation in this understudied area of precancerous lesions. PanINs are classified into PanIN 1, PanIN 2, and PanIN 3, with PanIN 3 having the highest likelihood of developing into invasive PDAC. Differentiating between PanIN 2 and PanIN 3 is clinically significant. Genetic alterations found in PDAC are also present in PanIN and increase with the grade of PanIN. Imaging methods alone are insufficient for distinguishing PanIN, necessitating the use of genetic and molecular tests for identification. In addition, metabolomics technologies and miRNAs are playing an increasingly important role in the field of cancer diagnosis, offering more possibilities for efficient identification of PanIN. Although detecting and stratifying the risk of PanIN poses challenges, the combined utilization of imaging, genetics, and metabolomics holds promise for improving patient survival in this field.
Collapse
Affiliation(s)
- Ling-ling Pian
- School of Medicine, The South China University of Technology, Guangzhou, Guangdong, China
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Mei-hui Song
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Teng-fei Wang
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Ling Qi
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Tie-li Peng
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Ke-ping Xie
- School of Medicine, The South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
31
|
Giurini EF, Ralph O, Pappas SG, Gupta KH. Looking Beyond Checkpoint Inhibitor Monotherapy: Uncovering New Frontiers for Pancreatic Cancer Immunotherapy. Mol Cancer Ther 2025; 24:18-32. [PMID: 39311547 DOI: 10.1158/1535-7163.mct-24-0311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/01/2024] [Accepted: 09/09/2024] [Indexed: 01/03/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) stands out as one of the most aggressive and challenging tumors, characterized by a bleak prognosis with a mere 11% survival rate over 5 years in the United States. Its formidable nature is primarily attributed to its highly aggressive behavior and poor response to existing therapies. PDAC, being notably resistant to immune interventions, presents a significant obstacle in treatment strategies. While immune checkpoint inhibitor therapies have revolutionized outcomes for various cancers, their efficacy in PDAC remains exceedingly low, benefiting less than 1% of patients. The consistent failure of these therapies in PDAC has prompted intensive investigation, particularly at the preclinical level, to unravel the intricate mechanisms of resistance inherent in this cancer type. This pursuit aims to pave the way for the development of novel immunotherapeutic strategies tailored to the distinct characteristics of PDAC. This review endeavors to provide a comprehensive exploration of these emerging immunotherapy approaches in PDAC, with a specific emphasis on elucidating their underlying immunological mechanisms. Additionally, it sheds light on the recently identified factors driving resistance to immunotherapy and evasion of the immune system in PDAC, offering insights beyond the conventional drivers that have been extensively studied.
Collapse
Affiliation(s)
- Eileena F Giurini
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
| | - Oliver Ralph
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
| | - Sam G Pappas
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
| | - Kajal H Gupta
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
- Division of Pediatric Surgery, Department of Surgery, Rush University Medical Center, Chicago, Illinois
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| |
Collapse
|
32
|
Hu Y, Liao Y, Pan S, Zhou J, Wan C, Huang F, Bai Y, Lin C, Xia Q, Liu Z, Gong J, Nie X, Wang M, Qin R. A Triple-Mismatch Differentiating assay exploiting activation and trans cleavage of CRISPR-Cas12a for mutation detection with ultra specificity and sensitivity. Biosens Bioelectron 2025; 267:116826. [PMID: 39369517 DOI: 10.1016/j.bios.2024.116826] [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: 09/01/2024] [Revised: 09/22/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Liquid biopsy technology is non-invasive and convenient, and is currently an emerging technology for cancer screening. Among them, clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR associated protein 12a (Cas12a) based nucleic acid detection technology has the advantages of high sensitivity, rapidity, and easy operation. However, CRISPR-Cas12a does not discriminate single-base mismatches of targets well enough to meet the needs of clinical detection. Herein, we developed the Triple-Mismatch Differentiating (TMD) assay. This assay amplified the small thermodynamic difference in mismatches at one site at the level of CRISPR-Cas12a activation to a significant thermodynamic difference at three sites at both the level of CRISPR-Cas12a activation and trans-cleavage, which greatly improves the ability of CRISPR-Cas12a to discriminate between base mismatches. Our manipulation greatly improved the specificity of the CRISPR-Cas12a system while maintaining its inherent sensitivity and simplicity, increasing the detection limit to 0.0001%. When testing samples from pancreatic cancer patients, our results were highly consistent with NGS sequencing results. We believe that the TMD assay will provide a new technology for early cancer detection and will be widely used in the clinical practice.
Collapse
Affiliation(s)
- Yibo Hu
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yangwei Liao
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Shutao Pan
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jingcong Zhou
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Changqing Wan
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Feiyang Huang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yu Bai
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Chen Lin
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Qilong Xia
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zixi Liu
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jun Gong
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Xiaoqi Nie
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
33
|
Fu X, Ma J, Ma F, Guo S, Wang X, Li Y, Tang Y, Qi J, Zhang W, Ye L. MISP-mediated enhancement of pancreatic cancer growth through the Wnt/β-catenin signaling pathway is suppressed by Fisetin. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167515. [PMID: 39278512 DOI: 10.1016/j.bbadis.2024.167515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
Pancreatic cancer is a highly malignant tumor characterized by high mortality and low survival rates. The mitotic interactor and substrate of Plk1 (MISP) is a cancer-associated protein that regulates mitotic spindle localization and is highly expressed in several malignant tumors, contributing to tumor development. However, the function and regulatory mechanisms of MISP in pancreatic cancer remain unclear. In this study, we analyzed RNA sequencing data related to pancreatic cancer from the TCGA and GEO databases, identifying MISP as a potential prognostic marker for the disease. MISP was significantly upregulated in pancreatic cancer cells and tissues compared to normal pancreatic cells and tissues. Notably, in pancreatic cancer cells, high MISP protein expression promoted cell proliferation and growth. Mechanistically, the upregulation of MISP facilitated the nuclear accumulation of β-catenin, thereby activating the Wnt/β-catenin signaling pathway and promoting pancreatic cancer growth. In search of effective inhibitors of MISP expression, we screened an FDA-approved drug library and identified Fisetin as a potential suppressor of MISP expression. Fisetin was found to downregulate the transcription factor MYB, thereby reducing MISP expression. Further experiments demonstrated that Fisetin effectively inhibited the in vitro and in vivo growth of pancreatic cancer by suppressing the MISP/Wnt/β-catenin signaling axis. In summary, our research has identified MISP as a novel therapeutic target in pancreatic cancer and uncovered its associated regulatory mechanisms.
Collapse
Affiliation(s)
- Xueli Fu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jiaqi Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Fangyuan Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shiman Guo
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xue Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ye Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yanxin Tang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jingwei Qi
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90001, USA
| | - Weiying Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| | - Lihong Ye
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
34
|
Zhang W, Chen J, Zhang W, Xu M. Advances in Endoscopic Ultrasound in Pancreatic Cancer Screening, Diagnosis, and Palliative Care. Biomedicines 2024; 13:76. [PMID: 39857661 PMCID: PMC11762820 DOI: 10.3390/biomedicines13010076] [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: 12/09/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy with a profoundly poor prognosis. Clinically, the condition most frequently manifests with symptoms including painless jaundice, abdominal discomfort, and back pain. Early diagnosis and the implementation of effective therapeutic strategies are critical for improving patient survival outcomes. However, merely 10-20% of patients are diagnosed at an early stage, with the majority presenting at advanced stages, often with metastasis. Consequently, early detection and intervention are crucial for enhancing prognosis. The widespread adoption of endoscopic ultrasonography (EUS) technology in recent years has significantly enhanced the diagnostic accuracy for pancreatic space-occupying lesions. EUS is increasingly recognized for its pivotal role in alleviating malignant biliary obstruction (MBO), gastric outlet obstruction (GOO), and refractory pain in advanced pancreatic cancer. This article aims to provide an overall review of the current applications of EUS in the diagnosis and treatment of pancreatic cancer, exploring its advantages and limitations in early screening, diagnosis, and palliative care. Furthermore, this review explores potential future directions in the field, aiming to provide valuable insights to inform and enhance the clinical management of pancreatic cancer.
Collapse
Affiliation(s)
- Wenyu Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Jingzheng Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Wei Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- Department of Gastroenterology, Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- Department of Gastroenterology, Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| |
Collapse
|
35
|
Liu C, Wu H, Cai D, Yu X. Unraveling the causal relationship between serum minerals and pancreatic cancer: a Mendelian randomization study. Discov Oncol 2024; 15:788. [PMID: 39692823 DOI: 10.1007/s12672-024-01695-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/11/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Pancreatic cancer is among the most lethal malignancies, characterized by a poor prognosis and limited modifiable factors. Emerging evidence indicates that serum mineral levels may influence the likelihood of developing pancreatic cancer. However, the causal relationship between serum minerals and pancreatic cancer remains unclear and warrants further investigation. METHODS This Mendelian randomization (MR) study was conducted to explore the causal effects of serum mineral levels on pancreatic cancer risk. Genetic variants associated with serum mineral levels, including calcium, iron, magnesium, zinc, selenium, and copper, were selected as instrumental variables (IVs) from large-scale genome-wide association study (GWAS) data. Multiple methods, including inverse variance weighting (IVW), MR-Egger, weighted median, weight methods, were employed to perform MR analysis. The effect sizes from the MR analysis, using two independent GWAS summary datasets related to pancreatic cancer, were combined through meta-analysis. The Cochrane Q test, MR-Egger intercept test, MR-PRESSO global test, and leave-one-out test were conducted for sensitivity tests. RESULTS Our MR analysis demonstrated a significant causal effect of genetically predicted serum calcium levels on increased pancreatic cancer risk [OR = 1.64, 95% CI 1.05-2.57, P = 0.029 (discovery cohort); OR = 1.52, 95% CI 1.07-2.15, P = 0.019 (validation cohort)], while no significant associations were found for other serum minerals (P > 0.05). Additional meta-analysis reinforces and substantiates this conclusion (pooled OR = 1.56, 95% CI 1.19-2.06, P = 0.001). No evidence of pleiotropy or heterogeneity was detected across multiple sensitivity tests (P > 0.05). CONCLUSION This study provides new evidence supporting the causal role of certain serum minerals, particularly calcium, in the development of pancreatic cancer. These findings may help inform future research into preventive strategies or therapies aimed at modulating mineral levels in patients at high risk of pancreatic cancer.
Collapse
Affiliation(s)
- Cong Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Huajun Wu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Dongdong Cai
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xin Yu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
36
|
Jiang X, Yuan Z, Ding T, Yu K, Dong J. SMS2 siRNA inhibits pancreatic tumor growth by tumor microenvironment modulation. Int Immunopharmacol 2024; 142:113111. [PMID: 39255679 DOI: 10.1016/j.intimp.2024.113111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 08/12/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024]
Abstract
The massive infiltration of suppressor immune cells within the tumor microenvironment (TME) of pancreatic ductal adenocarcinoma (PDAC) is a major cause of treatment resistance. Reducing this infiltration may represent a potentially effective therapeutic strategy. Sphingomyelin synthase 2 (SMS2) is a crucial enzyme for sphingomyelin synthesis, contributing significantly to the integrity and function of the plasma membrane. In this study, we developed a self-assembling SMS2 siRNA gene expression plasmid for in vivo delivery. The SMS2 siRNA specifically inhibits SMS2 expression while preserving the expression and activity of SMS1. Administration of the self-assembling SMS2 siRNA suppresses tumor growth in a murine model of Panc02 pancreatic carcinoma, modulates the polarization of tumor-associated macrophages (TAMs), and reduces the infiltration of tumor-associated neutrophils (TANs) by regulating the NF-κB/CXCL5 pathway. Consequently, utilizing SMS2 siRNA to improve the local immunosuppressive microenvironment holds promise for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Xin Jiang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, China
| | - Ziqing Yuan
- Experiment & Teaching Center, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Tingbo Ding
- Experiment & Teaching Center, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ker Yu
- Department of Pharmacology, School of Pharmacy, Fudan University, China.
| | - Jibin Dong
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, China.
| |
Collapse
|
37
|
Wu J, Tang L, Zheng F, Chen X, Li L. A review of the last decade: pancreatic cancer and type 2 diabetes. Arch Physiol Biochem 2024; 130:660-668. [PMID: 37646618 DOI: 10.1080/13813455.2023.2252204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/04/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
Pancreatic cancer (PC) is a prevalent gastrointestinal tumour known for its high degree of malignancy, resulting in a mere 10% five-year survival rate for most patients. Over the past decade, a growing body of research has shed light on the intricate bidirectional association between PC and Type 2 diabetes (T2DM). The collection of PC- and T2DM-related articles is derived from two comprehensive databases, namely WOS (Web of Science Core Collection) and CNKI (China National Knowledge Infrastructure). This article discusses the last 10 years of research trends in PC and T2DM and explores their potential regulatory relationship as well as related medications.
Collapse
Affiliation(s)
- Jiaqi Wu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Nursing, Southern Medical University, Guangzhou, China
| | - Liang Tang
- Department of General Medicine, Zhuzhou Central Hospital, Zhuzhou, China
| | - Feng Zheng
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Xun Chen
- Department of the Trauma center, Zhuzhou Central Hospital, Zhuzhou, China
- Department of hepatobiliary surgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Lei Li
- Department of Pathology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
38
|
Grabill N, Louis M, Cawthon M, Gherasim C, Chambers J. Diagnostic and therapeutic strategies in pancreatic adenosquamous carcinoma: Molecular and clinical insights in managing metastatic disease. Radiol Case Rep 2024; 19:6016-6026. [PMID: 39345848 PMCID: PMC11439409 DOI: 10.1016/j.radcr.2024.08.120] [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: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024] Open
Abstract
Adenosquamous carcinoma of the pancreas (ASCP) is a rare and aggressive variant of pancreatic cancer, characterized by both adenocarcinoma and squamous cell carcinoma components. It presents significant diagnostic and therapeutic challenges due to its atypical histology and poor prognosis. A 72-year-old male presented with abdominal pain, lighter-colored stools, and intermittent nausea. Initial imaging revealed a complex mass in the distal pancreatic body and tail. Elevated lipase levels and subsequent endoscopic ultrasound-guided fine-needle biopsy (EUS-FNB) suggested an atypical pancreatic lesion with keratinizing squamous cells. Further investigation through fiberoptic bronchoscopy and EBUS-guided transbronchial needle aspiration (TBNA) confirmed carcinoma with squamous differentiation. Genetic testing identified KRAS G12D and PIK3CA mutations. The multidisciplinary tumor board recommended systemic chemotherapy with mFOLFIRINOX and G-CSF support. The patient underwent twelve cycles of mFOLFIRINOX with dose adjustments for thrombocytopenia and effective management of chemotherapy-related side effects. Restaging CT scans showed a decrease in tumor size and stable metastatic nodes. The patient showed a partial biochemical response with decreasing CA 19-9 levels and disease stabilization on imaging. This case demonstrates the critical role of a multidisciplinary approach in managing rare pancreatic malignancies. ASCP requires a comprehensive diagnostic and therapeutic strategy involving advanced imaging, histopathological confirmation, and personalized chemotherapy. Integrating advanced diagnostic techniques, molecular profiling, and a multidisciplinary approach is essential for improving patient outcomes and providing comprehensive care for this challenging malignancy. Addressing the psychological aspects and offering compassionate care are vital for supporting patients through their treatment journey.
Collapse
Affiliation(s)
- Nathaniel Grabill
- Northeast Georgia Medical Center, General Surgery Department, Gainesville, GA 30501, USA
| | - Mena Louis
- Northeast Georgia Medical Center, General Surgery Department, Gainesville, GA 30501, USA
| | - Mariah Cawthon
- Northeast Georgia Medical Center, General Surgery Department, Gainesville, GA 30501, USA
| | - Claudia Gherasim
- Northeast Georgia Medical Center, Pathology Department, Gainesville, GA 30501, USA
| | - James Chambers
- Northeast Georgia Medical Center, General Surgery Department, Braselton, GA 30517, USA
| |
Collapse
|
39
|
Xiao M, Xue J, Jin E. SPOCK: Master regulator of malignant tumors (Review). Mol Med Rep 2024; 30:231. [PMID: 39392048 PMCID: PMC11487499 DOI: 10.3892/mmr.2024.13355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024] Open
Abstract
SPARC/osteonectin, CWCV and Kazal‑like domain proteoglycan (SPOCK) is a family of highly conserved multidomain proteins. In total, three such family members, SPOCK1, SPOCK2 and SPOCK3, constitute the majority of extracellular matrix glycoproteins. The SPOCK gene family has been demonstrated to serve key roles in tumor regulation by affecting MMPs, which accelerates the progression of cancer epithelial‑mesenchymal transition. In addition, they can regulate the cell cycle via overexpression, inhibit tumor cell proliferation by inactivating PI3K/AKT signaling and have been associated with numerous microRNAs that influence the expression of downstream genes. Therefore, the SPOCK gene family are potential cancer‑regulating genes. The present review summarizes the molecular structure, tissue distribution and biological function of the SPOCK family of proteins, in addition to its association with cancer. Furthermore, the present review documents the progress made in investigations into the role of SPOCK, whilst also discussing prospects for the future of SPOCK‑targeted therapy, to provide novel ideas for clinical application and treatment.
Collapse
Affiliation(s)
- Mingyuan Xiao
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110134, P.R. China
| | - Jiancheng Xue
- Department of Otolaryngology, Head and Neck Surgery, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
- Shenzhen Clinical Research Center for Otolaryngology Diseases, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
| | - Enli Jin
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110134, P.R. China
| |
Collapse
|
40
|
Yang W, Hu P, Zuo C. Application of imaging technology for the diagnosis of malignancy in the pancreaticobiliary duodenal junction (Review). Oncol Lett 2024; 28:596. [PMID: 39430731 PMCID: PMC11487531 DOI: 10.3892/ol.2024.14729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
The pancreaticobiliary duodenal junction (PBDJ) is the connecting area of the pancreatic duct, bile duct and duodenum. In a broad sense, it refers to a region formed by the head of the pancreas, the pancreatic segment of the common bile duct and the intraduodenal segment, the descending and the horizontal part of the duodenum, and the soft tissue around the pancreatic head. In a narrow sense, it refers to the anatomical Vater ampulla. Due to its complex and variable anatomical features, and the diversity of pathological changes, it is challenging to make an early diagnosis of malignancy at the PBDJ and define the histological type. The unique anatomical structure of this area may be the basis for the occurrence of malignant tumors. Therefore, understanding and subclassifying the anatomical configuration of the PBDJ is of great significance for the prevention and treatment of malignant tumors at their source. The present review comprehensively discusses commonly used imaging techniques and other new technologies for diagnosing malignancy at the PBDJ, offering evidence for physicians and patients to select appropriate examination methods.
Collapse
Affiliation(s)
- Wanyi Yang
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
- Graduates Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410013, P.R. China
| | - Pingsheng Hu
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
| | - Chaohui Zuo
- Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Province, Changsha, Hunan 410013, P.R. China
- Graduates Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
41
|
Khalid A, Faiz Z, Shah M, Newman E, King DA, DePeralta D, Gholami S, Weiss MJ, Melis M. Factors Influencing Immunotherapy Utilization in Stage IV Pancreatic Cancer: Impact of Race and Socioeconomics in the U.S. J Gastrointest Cancer 2024; 56:25. [PMID: 39592489 DOI: 10.1007/s12029-024-01119-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC), a highly lethal cancer with a poor prognosis, is expected to become the second deadliest cancer in the United States by 2030. Despite advancements in treatment modalities, the survival rates of patients with PDAC have remained low. Immunotherapy has emerged as a promising treatment for various cancers; however, its utilization in PDAC has been limited due to various challenges, including resistance mechanisms and the advanced stage at which most patients are diagnosed. METHODS We analyzed data from the National Cancer Database (NCDB) from 2010 to 2017, focusing on the impact of race, insurance status, and socioeconomic factors among patients with stage IV PDAC using logistic regression analyses. RESULTS Among 109,663 patients with stage IV PDAC, 421 (0.38%) received immunotherapy. The recipients were younger (median age 63 vs. 68 years, p < 0.001) and more likely to be white (87.4% vs. 82.1%). Patients with private insurance or Medicare (p < 0.001), and those earning more than $60 k annually (51.0% vs. 36.4%, p < 0.001) were more likely to receive immunotherapy. Treatment was more likely in academic/research programs than in community cancer programs (53.0% vs. 33.4%, p < 0.001). On multivariate analysis, Black patients had lower odds of receiving immunotherapy than Caucasian patients (OR: 0.74 [95% CI: 0.601-0.882], p = 0.019). Higher income was also a significant predictor of immunotherapy utilization (highest vs. lowest income quartile: OR, 2.228 [95% CI: 1.422-3.491], p < 0.001). CONCLUSIONS This study revealed significant disparities in immunotherapy access for stage IV PDAC based on race, socioeconomic status, and geographic location in the United States, highlighting the need for intervention to promote equitable access to this promising treatment modality.
Collapse
Affiliation(s)
- Abdullah Khalid
- Northwell Health, North Shore/Long Island Jewish General Surgery, 300 Community Dr., Manhasset, NY, USA.
| | - Zohaa Faiz
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Manav Shah
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, NY, USA
| | - Elliot Newman
- Northwell Health Lenox Hill Hospital, 100 E 77Th St, New York, NY, USA
| | - Daniel A King
- Northwell Health Cancer Institute, 1111 Marcus Ave, New Hyde Park, NY, USA
| | - Danielle DePeralta
- Northwell Health Cancer Institute, 1111 Marcus Ave, New Hyde Park, NY, USA
| | - Sepideh Gholami
- Northwell Health Cancer Institute, 1111 Marcus Ave, New Hyde Park, NY, USA
| | - Matthew J Weiss
- Northwell Health Cancer Institute, 1111 Marcus Ave, New Hyde Park, NY, USA
| | | |
Collapse
|
42
|
Fotros D, Tabar MS, Chegini M, Bahrizadeh M, Sadeghi A, Rabbani A, Yari Z, Hekmatdoost A. Adherence to the dietary approaches to stop hypertension (DASH) and risk of pancreatic steatosis. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2024; 43:190. [PMID: 39567983 PMCID: PMC11580558 DOI: 10.1186/s41043-024-00628-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 08/20/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND The Dietary Approach to Stop Hypertension (DASH) has shown positive effects on various health factors that may be related to pancreatic steatosis (PS). This study aimed to investigate the association between adherence to the DASH diet and the risk of developing PS. METHODS This case-control study was conducted on 278 patients diagnosed with gallstone disease and referred to Taleghani Hospital (Tehran, Iran). Among the participants, 89 were diagnosed with PS based on an endoscopic ultrasound (EUS) examination, while 189 patients did not exhibit this condition. The dietary intake of patients was assessed using a validated food frequency questionnaire (FFQ). Participants were classified based on the DASH diet score. Multiple logistic regression models estimated crude and multivariable-adjusted odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS The mean ± SD of DASH score in PS patients and controls was 23.68 ± 4.38 and 25.27 ± 4.2, respectively (P = 0.006). The risk of PS in the highest tertile of DASH score was 64% lower than the lowest tertile (OR = 0.36, 95%CI: 0.17-0.75, P = 0.005) after full adjustment for confounders. Also, more intake of vegetables and whole grains and less intake of sodium, red and processed meat were each significantly associated with reduced risk of PS. CONCLUSIONS Our data prove that adherence to the DASH diet was associated with a lower risk of PS. Further prospective studies are warranted to confirm these associations and explore the underlying mechanisms.
Collapse
Affiliation(s)
- Danial Fotros
- Department of Clinical Nutrition and dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Shaygan Tabar
- Department of Clinical Nutrition and dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maedeh Chegini
- Department of Clinical Nutrition and dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Bahrizadeh
- Department of Clinical Nutrition and dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Research Institute for Gastroenterology and Liver Diseases of Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhassan Rabbani
- Department of General Surgery, Ayatollah Taleghani Hospital, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Zahra Yari
- Department of Nutrition Research, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Azita Hekmatdoost
- Department of Clinical Nutrition and dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
43
|
Nopour R. Prediction of 12-month recurrence of pancreatic cancer using machine learning and prognostic factors. BMC Med Inform Decis Mak 2024; 24:339. [PMID: 39543603 PMCID: PMC11566389 DOI: 10.1186/s12911-024-02766-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND AND AIM Pancreatic cancer is lethal and prevalent among other cancer types. The recurrence of this tumor is high, especially in patients who did not receive adjuvant therapies. Early prediction of PC recurrence has a significant role in enhancing patients' prognosis and survival. So far, machine learning techniques have given us insight into favorable performance efficiency in various medical domains. So, this study aims to establish a prediction model based on machine learning to achieve better prediction on this topic. MATERIALS AND METHODS In this retrospective research, we used data from 585 PC patient cases from January 2019 to November 2023 from three clinical centers in Tehran City. Ten chosen ensemble and non-ensemble algorithms were used to establish prediction models on this topic. RESULTS Random forest and support vector machine with an AU-ROC of approximately 0.9 obtained more performance efficiency regarding PC recurrence. Lymph node metastasis, tumor size, tumor grade, radiotherapy, and chemotherapy were the best factors influencing PC recurrence. CONCLUSION Random forest and support vector machine algorithms demonstrated high-performance ability and clinical usability to improve doctors' decisions in achieving different therapeutic and diagnostic measures.
Collapse
Affiliation(s)
- Raoof Nopour
- Department of Health Information Management, Student Research Committee, School of Health Management and Information Sciences Branch, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
44
|
Küçükekmekci B, Budak Yıldıran FA. Investigation of the efficacy of siRNA-mediated KRAS gene silencing in pancreatic cancer therapy. PeerJ 2024; 12:e18214. [PMID: 39553720 PMCID: PMC11566511 DOI: 10.7717/peerj.18214] [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: 02/06/2024] [Accepted: 09/11/2024] [Indexed: 11/19/2024] Open
Abstract
Aim Pancreatic carcinoma is an aggressive cancer that progresses without many symptoms. The difficulty of early diagnosis and an inadequate response to traditional treatments also cause the survival rate of pancreatic cancer to be low. Current research is focusing on methods of diagnosis and treatment, such as gene therapy, to increase survival rates. Small interfering ribonucleic acid (siRNA) has emerged as a promising advanced therapeutic strategy for cancer treatment. This study sought to silence the KRAS gene in the human pancreatic carcinoma cell line using a complex of small interfering ribonucleic acid (siRNA) and gold nanoparticles (AuNP). Methods In this study, 25 nM siRNA and gold nanoparticles at 0.5 mg/ml, 0.25 mg/ml, and 0.125 mg/ml concentrations were used to silence the KRAS gene in the CAPAN-1 cell line. Real-time PCR analysis, agarose gel electrophoresis, and double staining were carried out, and xCelligence real-time cell analysis (RTCA) was used to measure proliferation. Results The PCR analysis revealed crossing point (CP) values of actin beta (ACTB) ranging from 33.04 to 35.98, which was in the expected range for all samples. The interaction between the gold nanoparticle/siRNA complex in the double staining analysis revealed that the most effective concentration of gold nanoparticle was 0.125 mg/ml. The WST-1 technique showed that siRNA/AuPEI cells in application groups had a viability rate of over 90%, indicating no toxicity or side effects. The xCELLigence RTCA® showed that at hour 72, there was a significant difference in proliferation in the 0.5 mg/mL PEI/AuNP-siRNA, 0.25 mg/mL PEI/AuNP-siRNA, and 0.125 mg/mL PEI/AuNP-siRNA application groups compared to the control and siRNA groups (p < 0.05). By hour 96, all three groups were statistically different from the control and siRNA groups in terms of proliferation (p < 0.05). Conclusions The results of this analysis suggest that the AuPEI/siRNA complex can be effectively used to silence the target gene, but more studies are needed to verify these results.
Collapse
Affiliation(s)
- Büşra Küçükekmekci
- Institute of Sciences, Department of Biology, Kırıkkale University, Kırıkkale, Yahşihan, Turkey
| | - Fatma Azize Budak Yıldıran
- Vocational High School of Health Care Services, Department of Medical Services and Techniques, Kırıkkale University, Kırıkkale, Yahşihan, Turkey
| |
Collapse
|
45
|
Khatkov IE, Maev IV, Bordin DS, Kononenko IB, Kucheryavyy YA, Pokataev IA, Snegovoy AV, Tryakin AA, Feoktistova PS, Zhukova LG. Role of enzyme replacement therapy for exocrine and nutritional insufficiency in patients with malignancies: A review. JOURNAL OF MODERN ONCOLOGY 2024; 26:380-389. [DOI: 10.26442/18151434.2024.3.203007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Exocrine pancreatic insufficiency (EPI) is a condition in which the amount of secreted pancreatic enzymes is insufficient to maintain normal digestion. EPI is a frequent complication of pancreatic or other malignancies. The presence of EPI in a cancer patient may be suggested by symptoms of maldigestion, malabsorption, and alteration of nutritional markers; however, it is important to note that the EPI symptoms may be subtle. In the early stages, EPI may be latent and manifested by malnutrition. However, even in the later stages, the symptoms of EPI may be similar to those of cancer or be masked by the condition after chemoradiation therapy. Antitumor therapy itself may also cause EPI. Enzyme replacement therapy (ERT) is the standard of care for EPI, but it is rarely prescribed to cancer patients. However, supportive therapy plays an essential role in treating cancer patients because the quality of life and life expectancy of patients largely depend on the adequacy of the complex treatment. The review discusses the possible causes of EPI and its diagnosis and treatment in cancer patients. Special attention is paid to ERT regimens, including those for improving digestion and the drug's dosage form. It is shown that pancreatin in minimicrospheres is the drug of choice for ERT, since the minimum particle size facilitates the most physiological digestion process.
Collapse
Affiliation(s)
- Igor E. Khatkov
- Loginov Moscow Clinical Scientific Center
- Russian University of Medicine
| | | | - Dmitry S. Bordin
- Loginov Moscow Clinical Scientific Center
- Russian University of Medicine
- Tver State Medical University
| | - Inessa B. Kononenko
- Lopatkin Scientific Research Institute of Urology and Interventional Radiology – Branch of National Medical Research Radiological Centre
| | | | - Ilya A. Pokataev
- Moscow City Hospital named after S.S. Yudin, Moscow Healthcare Department
| | - Anton V. Snegovoy
- Russian University of Medicine
- Lopatkin Scientific Research Institute of Urology and Interventional Radiology – Branch of National Medical Research Radiological Centre
| | | | - Polina S. Feoktistova
- Loginov Moscow Clinical Scientific Center
- Central State Medical Academy of the President of the Russian Federation
| | | |
Collapse
|
46
|
González Abós C, Lorenzo C, Archilla I, Vidal-Robau N, Cuatrecasas M, Ausania F. Effect of warm and cold ischemia on pancreaticoduodenectomy specimen following robotic pancreaticoduodenectomy. BMC Surg 2024; 24:346. [PMID: 39501227 PMCID: PMC11536913 DOI: 10.1186/s12893-024-02652-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/30/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND The adoption of robotic pancreaticoduodenectomy (PD) has increased in recent years for the treatment of pancreatic head tumors and periampullary lesions. Some potential benefits seem to be demonstrated; however, obtaining specimens through this method can potentially compromise the diagnosis depending on the timing of the specimen retrieval, and the impact of longer perioperative time on ischemia and autolysis of the surgical specimen has not been analyzed. The aim of this study is to evaluate the histological changes associated with timing of specimen retrieval during robotic PD. METHODS A review of histopathology files was performed for all pancreatic specimens collected at our hospital from January 2022 to March 2024. Both warm ischemia time (WIT) and cold ischemia time (CIT) were collected. Histological features related to ischemic damage were evaluated in normal duodenal and pancreatic parenchyma as well as pancreatic tumor, and were graded as: absent, mild, moderate and severe. Univariate and multivariate analyses were performed to determine which variables were associated with moderate and severe ischemic changes. RESULTS Sixty surgical specimens were analyzed: 20 open PD, 17 robotic PD with cold ischemia, and 23 robotic PD. Median total WIT was 182 min (open PD 57 min vs. RPD 190 min vs. RPD-CI 198 min; p < 0.001). Median CIT was 760 min (740-835) in samples stored at 4ºC. Univariate analysis showed that longer intraoperative time, male gender and cold ischemia were associated with pancreatic tissue ischemic changes. In multivariate analysis, cold ischemia was the only independent factor associated with normal pancreatic tissue and tumor tissue moderate and severe ischemic changes. CONCLUSIONS Prolonged ischemia time, especially in the case of cold storage, has a strong effect on the degradation of normal and tumor tissue without affecting pathological evaluation. Operative teams should aim to optimize both the duration and efficiency of the surgical procedure, ensuring minimal ischemic time. Simultaneously, pathology departments must be equipped to process pancreatic specimens promptly, with protocols in place to minimize the time between resection and analysis.
Collapse
Affiliation(s)
- Carolina González Abós
- Department of HBP and Transplant Surgery, Hospital Clinic de Barcelona, C/ Villarroel 170, Barcelona, 08036, Spain.
- University of Barcelona, Barcelona, Spain.
- Gene Therapy and Cancer, IDIBAPS, Barcelona, Spain.
| | - Claudia Lorenzo
- Department of HBP and Transplant Surgery, Hospital Clinic de Barcelona, C/ Villarroel 170, Barcelona, 08036, Spain
| | - Iván Archilla
- Department of Pathology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Nuria Vidal-Robau
- Department of Pathology, Hospital Clinic de Barcelona, Barcelona, Spain
| | | | - Fabio Ausania
- Department of HBP and Transplant Surgery, Hospital Clinic de Barcelona, C/ Villarroel 170, Barcelona, 08036, Spain
- University of Barcelona, Barcelona, Spain
- Gene Therapy and Cancer, IDIBAPS, Barcelona, Spain
| |
Collapse
|
47
|
Lencioni G, Gregori A, Toledo B, Rebelo R, Immordino B, Amrutkar M, Xavier CPR, Kocijančič A, Pandey DP, Perán M, Castaño JP, Walsh N, Giovannetti E. Unravelling the complexities of resistance mechanism in pancreatic cancer: Insights from in vitro and ex-vivo model systems. Semin Cancer Biol 2024; 106-107:217-233. [PMID: 39299411 DOI: 10.1016/j.semcancer.2024.09.002] [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: 07/19/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with poor prognosis and rising global deaths. Late diagnosis, due to absent early symptoms and biomarkers, limits treatment mainly to chemotherapy, which soon encounters resistance. PDAC treatment innovation is hampered by its complex and heterogeneous resistant nature, including mutations in key genes and a stromal-rich, immunosuppressive tumour microenvironment. Recent studies on PDAC resistance stress the need for suitable in vitro and ex vivo models to replicate its complex molecular and microenvironmental landscape. This review summarises advances in these models, which can aid in combating chemoresistance and serve as platforms for discovering new therapeutics. Immortalised cell lines offer homogeneity, unlimited proliferation, and reproducibility, but while many gemcitabine-resistant PDAC cell lines exist, fewer models are available for resistance to other drugs. Organoids from PDAC patients show promise in mimicking tumour heterogeneity and chemosensitivity. Bioreactors, co-culture systems and organotypic slices, incorporating stromal and immune cells, are being developed to understand tumour-stroma interactions and the tumour microenvironment's role in drug resistance. Lastly, another innovative approach is three-dimensional bioprinting, which creates tissue-like structures resembling PDAC architecture, allowing for drug screening. These advanced models can guide researchers in selecting optimal in vitro tests, potentially improving therapeutic strategies and patient outcomes.
Collapse
Affiliation(s)
- Giulia Lencioni
- Fondazione Pisana per La Scienza, San Giuliano Terme, Italy; Department of Biology, University of Pisa, Pisa, Italy
| | - Alessandro Gregori
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Belén Toledo
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain
| | - Rita Rebelo
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto 4200-135, Portugal; Cancer Drug Resistance Group, Institute of Molecular Pathology and Immunology (IPATIMUP), University of Porto, Porto 4200-135, Portugal; Department of Biological Sciences, Faculty of Pharmacy of the University of Porto (FFUP), Porto, Portugal
| | - Benoît Immordino
- Fondazione Pisana per La Scienza, San Giuliano Terme, Italy; Institute of Life Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Manoj Amrutkar
- Department of Pathology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Cristina P R Xavier
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto 4200-135, Portugal; Cancer Drug Resistance Group, Institute of Molecular Pathology and Immunology (IPATIMUP), University of Porto, Porto 4200-135, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, Gandra, Portugal
| | - Anja Kocijančič
- Centre for Embryology and Healthy Development, Department of Microbiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Deo Prakash Pandey
- Centre for Embryology and Healthy Development, Department of Microbiology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain
| | - Justo P Castaño
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain; Reina Sofia University Hospital, Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Naomi Walsh
- Life Sciences Institute, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Elisa Giovannetti
- Fondazione Pisana per La Scienza, San Giuliano Terme, Italy; Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands; Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
48
|
Maurer E, Lehman B, Matthäi E, Denzer U, Figiel J, Jesinghaus M, Slater EP, Stefenelli U, Gress TM, Bartsch DK. Pancreatic cancer screening is effective in individuals at risk with predisposing germline gene variants, but not in gene variant-negative familial pancreatic cancer families. United European Gastroenterol J 2024; 12:1211-1221. [PMID: 39031472 PMCID: PMC11578844 DOI: 10.1002/ueg2.12631] [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: 03/17/2024] [Accepted: 06/20/2024] [Indexed: 07/22/2024] Open
Abstract
OBJECTIVE To evaluate the diagnostic yield of pancreatic cancer screening in individuals at risk (IAR) from familial pancreatic cancer (FPC) families with respect to the presence or absence of pathogenic germline variants predisposing to pancreatic adenocarcinoma (PDAC). DESIGN In a 20 years period, IAR from FPC families were enrolled in a prospective screening program of the national case collection for FPC of Germany, including magnet resonance imaging (MRI) and endoscopic ultrasound (EUS). The diagnostic yield was analyzed regarding significant pancreatic lesions such as PDAC, high-grade pancreatic-intraepithelial-neoplasia (PanIN3) and intraductal-papillary-mucinous-neoplasia (IPMN) with high-grade dysplasia. Screening results were compared between carriers of pathogenic variants and variant-negative IAR. RESULTS 337 IAR, including 74 (22%) variant-carriers and 263 IAR of variant-negative FPC families (mean age 49; standard deviation [SD] + 8.9) were followed 64 (SD + 55) months. IAR underwent 5.1 (SD + 3.9) screening visits with 1733 MRI (5.1,SD + 3.9 per IAR) and 728 EUS (2.2,SD + 1.7 per IAR). In 12 (4%) cases, significant pancreatic lesions were detected, including 4 PDAC, 3 PanIN3 and 5 high-grade IPMN. Three of 4 IAR with PDAC died after a mean of 27 months postoperatively, and one IAR is alive without evidence of disease after 31 months. The diagnostic yield for significant lesions was 13.5% (10/74) for variant carriers compared to 0.8% (2/263) for IAR of variant-negative FPC families (p < 0.001). Logistic regression analysis revealed that a negative variant status was almost always accompanied by the absence of a significant lesion over time with a negative predictive value of 99.2% (95% CI 97.3%-99.9%). CONCLUSION The diagnostic yield seems to justify PDAC screening in IAR of FPC-families with pathogenic germline variants in PDAC predisposing genes, not in IAR of variant-negative families.
Collapse
Affiliation(s)
- Elisabeth Maurer
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| | - Bettina Lehman
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| | - Elvira Matthäi
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| | - Ulrike Denzer
- Department of GastroenterologyEndocrinologyMetabolism and InfectiologyPhilipps University MarburgMarburgGermany
| | - Jens Figiel
- Department of Diagnostic and Interventional RadiologyPhilipps University MarburgMarburgGermany
| | | | - Emily P. Slater
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| | | | - Thomas M. Gress
- Department of GastroenterologyEndocrinologyMetabolism and InfectiologyPhilipps University MarburgMarburgGermany
| | - Detlef K. Bartsch
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| |
Collapse
|
49
|
Limbu KR, Chhetri RB, Kim S, Shrestha J, Oh YS, Baek DJ, Park EY. Targeting sphingosine 1-phosphate and sphingosine kinases in pancreatic cancer: mechanisms and therapeutic potential. Cancer Cell Int 2024; 24:353. [PMID: 39462385 PMCID: PMC11514880 DOI: 10.1186/s12935-024-03535-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
Pancreatic cancer is known to be the most lethal cancer. Fewer new treatments are being developed for pancreatic cancer as compared to other cancers. The bioactive lipid S1P, which is mainly regulated by sphingosine kinase 1 (SK1) and sphingosine kinase 2 (SK2) enzymes, plays significant roles in pancreatic cancer initiation and exacerbation. S1P controls many signaling pathways to modulate the progression of pancreatic cancer through the G-coupled receptor S1PR1-5. Several papers reporting amelioration of pancreatic cancer via modulation of S1P levels or downstream signaling pathways have previously been published. In this paper, for the first time, we have reviewed the results of previous studies to understand how S1P and its receptors contribute to the development of pancreatic cancer, and whether S1P can be a therapeutic target. In addition, we have also reviewed papers dealing with the effects of SK1 and SK2, which are kinases that regulate the level of S1P, on the pathogenesis of pancreatic cancer. We have also listed available drugs that particularly focus on S1P, S1PRs, SK1, and SK2 for the treatment of pancreatic cancer. Through this review, we would like to suggest that the SK/S1P/S1PR signaling system can be an important target for treating pancreatic cancer, where a new treatment target is desperately warranted.
Collapse
Affiliation(s)
- Khem Raj Limbu
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea
| | | | - Subin Kim
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea
| | - Jitendra Shrestha
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA
| | - Yoon Sin Oh
- Department of Food and Nutrition, Eulji University, Seongnam, 13135, South Korea
| | - Dong Jae Baek
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea.
| | - Eun-Young Park
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea.
| |
Collapse
|
50
|
Groß T, Merboth F, Klimowa A, Kahlert C, Distler M, Weitz J, Welsch T, Müssle B. Impact of an enhanced anti-infection prophylaxis strategy for pancreatoduodenectomy: a single centre analysis. Langenbecks Arch Surg 2024; 409:307. [PMID: 39402424 PMCID: PMC11473572 DOI: 10.1007/s00423-024-03465-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/01/2024] [Indexed: 10/19/2024]
Abstract
INTRODUCTION : Surgical site infection (SSI) after pancreatoduodenectomy (PD) is a significant concern. Targeted antibiotic prophylaxis (pAP) has been tested to mitigate antibiotic resistance patterns, especially after preoperative bile duct stenting. The aim of this study was to investigate the effect of enhanced anti-infective prophylaxis (EAP) on the incidence of superficial and intraabdominal SSI. METHODS All patients who underwent PD at a single centre between May 2018 and May 2021 were retrospectively analysed. A control cohort of patients who received pAP with intravenous cefuroxime and metronidazole and routine intraoperative abdominal lavage according to the surgeons' preferences. Since March 2020, pAP has been changed to piperacillin/tazobactam according to local resistance patterns and combined with routine intraoperative extended abdominal lavage (EIPL). Preoperative selective decontamination of the digestive tract (SDD) has been applied routinely since Jan 2019. RESULTS In total, 163 patients were included. The standard (n = 100) and EAP (n = 63) groups did not significantly differ with regard to pertinent patient and operative characteristics. In the EAP group, the rates of SSI (14% vs. 37%, p = 0.002, total rate: 28%) and urinary tract infection (24% vs. 8%, p = 0.011, total rate 18%) were significantly lower. Other septic complications were not significantly different. In addition, the risk of developing gastrointestinal bleeding and delayed gastric emptying was significantly lower in the EAP group. Multivariate analysis showed that an age > 67 years was a significant risk factor for SSI. CONCLUSION The results indicate that enhanced anti-infective prophylaxis may significantly decrease the incidence of SSI in patients after PD.
Collapse
Affiliation(s)
- Tina Groß
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Ependorf, Hamburg, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Felix Merboth
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
| | - Anna Klimowa
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
| | - Christoph Kahlert
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidberg, Heidelberg, Germany
| | - Marius Distler
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Thilo Welsch
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Ependorf, Hamburg, Germany
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Benjamin Müssle
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- Department of General and Visceral Surgery, University Hospital Ulm, Ulm, Germany.
| |
Collapse
|