1
|
Zhang L, Yuan J, Yao S, Wen G, An J, Jin H, Tuo B. Role of m5C methylation in digestive system tumors (Review). Mol Med Rep 2025; 31:142. [PMID: 40183387 PMCID: PMC11979572 DOI: 10.3892/mmr.2025.13507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Currently, the incidence of digestive system tumors has been increasing annually, thus becoming a prevalent cause of cancer‑related mortalities. Although significant strides have been made in targeting the molecular mechanisms that underpin the development of these tumors, their treatment and prognosis still pose substantial challenges. This is primarily due to the ambiguity of early diagnostic indicators and the fact that most digestive system tumors are detected at an advanced stage. However, epigenetic modifications are capable of altering the expression of oncogenes and regulating biological processes in cancer. In recent years, the study of methylation in relation to tumor pathogenesis has become a focus of prominent research. Among the various types of methylation, 5‑methylcytosine (m5C) methylation plays a crucial role in the development of digestive system tumors and is anticipated to serve as a novel therapeutic target. However, to date, a comprehensive and systematic review concerning the role of m5C methylation in digestive system tumors is lacking. Consequently, the present study reviewed the role of m5C methylation in digestive system tumors such as esophageal cancer, gastric cancer and hepatocellular carcinoma, with the aim of providing a valuable reference for future research endeavors.
Collapse
Affiliation(s)
- Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jianbo Yuan
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, P.R. China
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Guorong Wen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jiaxing An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
2
|
Wong P, Tran TB, Pollini T, Hernandez S, Zampese M, Todeschini L, Aguilar LL, Maker VK, Maker AV. Impact of coronary artery stenting on perioperative mortality and complications in patients undergoing pancreaticoduodenectomy. J Gastrointest Surg 2025; 29:102020. [PMID: 40118202 DOI: 10.1016/j.gassur.2025.102020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/03/2025] [Accepted: 03/15/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Postoperative morbidity and mortality rates from pancreaticoduodenectomy (PD) have significantly decreased, allowing for greater consideration of patients with severe comorbidities. This study aimed to evaluate the effect of previous coronary artery intervention on morbidity and mortality among patients who underwent PD. METHODS Patients who underwent PD were identified from the American College of Surgeon National Surgical Quality Improvement Program database. Patients with previous coronary artery intervention received either balloon dilatation or stent placement. The main outcome measures included in-hospital mortality and postoperative myocardial infarction (MI). RESULTS Of 10,848 patients who underwent PD, 698 (6.4%) received previous coronary artery intervention. Compared with patients without coronary artery intervention, those with previous coronary artery intervention were older (65 vs 70 years, respectively; P <.001), were less likely to be female (50.2% vs 26.4%, respectively; P <.001), and had higher median body mass index (26 vs 27 kg/m2, respectively; P =.003). Compared with patients not in the angioplasty/stent cohort, those in the angioplasty/stent cohort were more likely to have diabetes mellitus (22.0% vs 39.3%, respectively), functional impairment (2.4% vs 4.9%, respectively), chronic obstructive pulmonary disease (4.1% vs 8.2%, respectively), hypertension (51.2% vs 86.2%, respectively), and bleeding disorders (2.2% vs 8.0%, respectively) (all P <.001). Compared with patients not in the angioplasty/stent cohort, those in the stent/angioplasty group were more likely to have postoperative complications (41.0% vs 51.4%, respectively; P <.001). Previous stent/angioplasty procedure (odds ratio [OR], 2.61 [95% CI, 1.42-4.57]; P =.001) was associated with developing postoperative MI but was not an independent predictor of in-hospital mortality (OR, 1.19 [95% CI, 0.81-1.70]; P =.369). CONCLUSION Previous stent placement/angioplasty was not associated with increased in-hospital mortality in patients who underwent PD, despite being correlated with an increased risk of MI and severe complications. Previous coronary artery angioplasty and/or stenting is not an absolute contraindication for PD, but patients should be medically optimized preoperatively to mitigate the risk of major adverse cardiac events.
Collapse
Affiliation(s)
- Paul Wong
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Thuy B Tran
- Division of Surgical Oncology, Department of Surgery, University of Illinois Chicago, Chicago, IL, United States
| | - Tommaso Pollini
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Sophia Hernandez
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Marco Zampese
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Letizia Todeschini
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Luis Laurean Aguilar
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Vijay K Maker
- Division of Surgical Oncology, Department of Surgery, University of Illinois Chicago, Chicago, IL, United States
| | - Ajay V Maker
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
3
|
Trieu J, Gilman A, Konstantinoff K, Lozano MD, Saieg M. Cytology-Radiology Correlation Series: Pancreatic cytopathology. Cancer Cytopathol 2025; 133:e70012. [PMID: 40232932 PMCID: PMC11999058 DOI: 10.1002/cncy.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/03/2025] [Accepted: 02/17/2025] [Indexed: 04/17/2025]
Abstract
The prevalence of pancreatic lesions has increased over the years because of an increase in accessibility to and the quality of cross-sectional imaging. This commentary describes the common non-neoplastic and neoplastic pancreatic lesions. The images in this commentary depict classic cross-sectional images, sonographic findings, and the cytopathologic diagnosis of each lesion. Most common non-neoplastic lesions include pseudocysts, autoimmune pancreatitis, and chronic pancreatitis. Most common neoplastic lesions include serous cystadenomas, intraductal papillary mucinous neoplasms, mucinous cystic neoplasms, solid pseudopapillary neoplasms, neuroendocrine tumors, pancreatic ductal adenocarcinoma, acinar cell carcinoma, and metastases to the pancreas. The aim of this Cytoimaging Correlation Series is to demonstrate the multidisciplinary involvement in the diagnosis of pancreatic pathology and to highlight main findings in the most common entities found in everyday practice.
Collapse
Affiliation(s)
- Judy Trieu
- Division of GastroenterologyWashington University School of MedicineSt LouisMissouriUSA
| | - Andrew Gilman
- Division of Gastroenterology and HepatologyUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Katerina Konstantinoff
- Mallinckrodt Institute of RadiologyWashington University School of MedicineSt LouisMissouriUSA
| | - Maria D. Lozano
- Department of PathologyUniversity of Navarra‐PamplonaPamplonaSpain
| | - Mauro Saieg
- Department of PathologyJewish General HospitalMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
4
|
Yamane K, Anazawa T, Nagai K, Kasai Y, Masui T, Izuwa A, Kurahashi K, Ishida S, Ogiso S, Yoshimura M, Iwai T, Matsubara J, Fukuda A, Isoda H, Hidaka Y, Ibi Y, Hatano E. Neoadjuvant Chemoradiotherapy Using Moderately Hypofractionated Intensity-Modulated Radiotherapy Versus Upfront Surgery for Resectable Pancreatic Cancer: A Retrospective Cohort Study. Ann Surg Oncol 2025; 32:3603-3613. [PMID: 39893341 PMCID: PMC11976822 DOI: 10.1245/s10434-025-16956-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/16/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND The efficacy of neoadjuvant chemoradiotherapy for resectable pancreatic ductal adenocarcinoma (R-PDAC) remains unclear. This study was designed to evaluate neoadjuvant chemoradiotherapy by using intensity-modulated radiotherapy (NAC-IMRT) for R-PDAC compared with upfront surgery (UpS). METHODS Among 198 patients with R-PDAC who were indicated for resection between 2013 and 2021, 130 were included in this study after excluding patients who underwent neoadjuvant chemotherapy and did not meet the NAC-IMRT criteria (Eligible set). NAC-IMRT was planned for 58 patients, and UpS was planned for 72 patients. Additionally, in 105 patients who could undergo the planned treatment (As-treated set), the surgical, pathological, and oncological outcomes were evaluated. RESULTS In the Eligible set, median overall survival (OS) was 50.5 months with NAC-IMRT and 34.7 months with UpS and progression-free survival was 20.4 months with NAC-IMRT and 13.9 months with UpS. In the As-treated set, OS was longer in the NAC-IMRT group (66.7 months vs. 34.7 months, p = 0.007). On multivariate analysis, NAC-IMRT was identified as an independent factor for better OS (hazard ratio 0.617, 95% confidence interval 0.382-0.995, p = 0.047, in the Eligible set). The incidence of postoperative complications did not show a difference between the two groups, and NAC-IMRT suppressed local tumor invasion, including lymphatic, venous, perineural invasion, and lymph node metastases. CONCLUSIONS NAC-IMRT may offer superior survival outcomes and manageable toxicity in R-PDAC patients compared with upfront surgery. This study supports the efficacy and safety of NAC-IMRT and recommends its consideration in R-PDAC treatment protocols.
Collapse
Affiliation(s)
- Kei Yamane
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takayuki Anazawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Kazuyuki Nagai
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yosuke Kasai
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshihiko Masui
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Surgery, Kurashiki Central Hospital, Okayama, Japan
| | - Aya Izuwa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koki Kurahashi
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Ishida
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Ogiso
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Michio Yoshimura
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiro Iwai
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junichi Matsubara
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyoshi Isoda
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yu Hidaka
- Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yumiko Ibi
- Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Etsuro Hatano
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
5
|
Arseneau RJ, Kempster E, Bekkers C, Samson T, Gala-Lopez BL, Ramjeesingh R, Boudreau JE, Arnason T. Claudin 18 (43-14A clone) expression in pancreatic ductal adenocarcinoma: Assessment of a potential clinical biomarker for zolbetuximab therapy. Transl Oncol 2025; 55:102362. [PMID: 40117781 PMCID: PMC11979426 DOI: 10.1016/j.tranon.2025.102362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/08/2025] [Accepted: 03/16/2025] [Indexed: 03/23/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains highly lethal, with a five-year survival rate below 15 %. Claudin 18.2 (CLDN18.2) has emerged as a novel potential therapeutic target in PDAC. Zolbetuximab, a monoclonal antibody targeting CLDN18.2, has demonstrated therapeutic benefit in gastric cancers and is now in phase 2 clinical trials for PDAC. Trial eligibility for zolbetuximab requires tumor membranous immunohistochemical staining with the pan-claudin 18 companion diagnostic antibody clone 43-14A. However, the expression of CLDN18 detected using this clone has only been evaluated in 62 patients from a single retrospective study. Herein, we report immunohistochemical staining using 43-14A on surgically resected PDAC samples (n = 120). Samples were stained following the protocol used in clinical trials, using the 43-14A VENTANA antibody in a prediluted kit, and according to the manufacturer's recommended protocol. Positive cases were defined as ≥ 75 % of tumor cells exhibiting membranous staining with an intensity of ≥ 2+. Out of 120 PDAC cases, 39 (32.5 %) stained positive for CLDN18 with 43-14A. A significant association was observed between lower tumor grade and higher 43-14A staining (p < 0.05). CLDN18-positive cases demonstrated significantly improved survival at the cohort's median overall survival (23 months, p < 0.05), suggesting that claudin expression could serve as a both a diagnostic and prognostic marker. Our findings indicate that 32.5 % of PDAC tumors in this cohort are positive for CLDN18, suggesting that a significant proportion of patients with PDAC could benefit from zolbetuximab and other CLDN18.2 targeted immunotherapies if pancreatic cancer therapeutic trials prove successful.
Collapse
Affiliation(s)
- Riley J Arseneau
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | | | | | | | - Boris L Gala-Lopez
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada; Nova Scotia Health, Halifax, NS, Canada; Department of Microbiology & Immunology, Dalhousie University, NS, Canada
| | - Ravi Ramjeesingh
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada; Nova Scotia Health, Halifax, NS, Canada
| | - Jeanette E Boudreau
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada; Nova Scotia Health, Halifax, NS, Canada; Department of Microbiology & Immunology, Dalhousie University, NS, Canada
| | - Thomas Arnason
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Nova Scotia Health, Halifax, NS, Canada.
| |
Collapse
|
6
|
Lee JC, Kang SW, Sim EJ, Bae JS, Koo SM, Byoun MS, Kwon S, Hong S, Kim Y, Youn Y, Jung K, Kim J, Jeong HH, Kim J, Hwang JH. Novel mRNA biomarker-based liquid biopsy for the detection of resectable pancreatic cancer. BMC Cancer 2025; 25:762. [PMID: 40269781 PMCID: PMC12016232 DOI: 10.1186/s12885-025-14124-w] [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: 09/19/2024] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies and most often diagnosed at an advanced stage. Identification of markers for the early diagnosis of PDAC is crucial. In this study, we aimed to identify novel mRNA biomarkers for diagnosing PDAC, focusing on early-stage tumorigenesis and associated immunological changes. METHODS Blood samples and clinical information from 1,963 individuals were obtained from a single tertiary hospital between 2015 and 2021. Candidate mRNA biomarkers were identified through literature review, and their expression levels in buffy coat samples were measured using reverse-transcription quantitative polymerase chain reaction. Machine learning-based feature selection confirmed the final biomarker panel, which was tested using an independent dataset for diagnostic performance. RESULTS In total, 1,504 individuals (417 patients with PDAC and 1,087 non-disease controls) were eligible for the study. Among the 55 candidate biomarkers identified, 15 mRNAs (CCL5, CCR5, CLEC7A, CXCL8, CXCR2, CXCR4, FOXP3, IFNA1, IFNL1, PTGES, PTGES2, PTGS2, SLC27A2, TNF, and VEGFA) were selected based on their diagnostic performance in distinguishing PDAC from control groups. The final model, HELP-15 (Human Early Liquid biopsy for PDAC), identified all PDAC stages (area under the curve [AUC] = 0.956) in the test set. For resectable pancreatic cancer (RPC), the AUC was 0.968, compared to 0.910 for carbohydrate antigen 19 - 9 (CA19-9). The combined model of the panel and CA19-9 achieved an AUC of 0.985 in patients with RPC. For all PDAC stages in patients with normal CA19-9 levels, the AUC of the panel was 0.967, whereas CA19-9 alone or in combination with the panel had AUCs of 0.658 and 0.885, respectively. CONCLUSION Compared to CA19-9, the mRNA biomarker panel, HELP-15, improved diagnostic performance in patients with RPC, particularly in those with normal CA19-9 levels.
Collapse
Affiliation(s)
- Jong-Chan Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Won Kang
- Research Center, HuVet bio Inc, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Eun-Jin Sim
- Research Center, HuVet bio Inc, Seoul, Korea
| | - Jin-Sik Bae
- Research Center, HuVet bio Inc, Seoul, Korea
| | | | | | - Serin Kwon
- Department of Life Science, Dongguk University, Seoul, Korea
| | - Seoi Hong
- Department of Computer and Artificial Intelligence, Dongguk University, Seoul, Korea
| | - Yunji Kim
- Department of Computer and Artificial Intelligence, Dongguk University, Seoul, Korea
| | - Yuna Youn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kwangrok Jung
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | | | - Jihie Kim
- Department of Computer and Artificial Intelligence, Dongguk University, Seoul, Korea.
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
7
|
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
|
8
|
Zare-Mehrjardi MJ, Hatami-Araghi M, Jafari-Khorchani M, Oushyani Roudsari Z, Taheri-Anganeh M, Abdolrahmat M, Ghasemi H, Aiiashi S. RNA biosensors for detection of pancreatic cancer. Clin Chim Acta 2025; 571:120237. [PMID: 40081786 DOI: 10.1016/j.cca.2025.120237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Pancreatic cancer is recognized as one of the most lethal types of cancer globally, characterized by a high mortality rate and a bleak prognosis, which greatly contributes to cancer-related deaths. Forecasts suggest that by 2030, pancreatic cancer will exceed other cancer types in prevalence. The disease presents considerable difficulties owing to the lack of prominent symptoms in its early stages, restricted options for early detection, rapid progression, and unfavorable outcomes. Presently, traditional methods for diagnosing pancreatic cancer primarily rely on imaging techniques. However, these methods often entail significant costs, require considerable time, and necessitate specialized skills for both operating the equipment and interpreting the resulting images. To overcome these obstacles, the use of biosensors has been proposed as a potentially valuable tool for the early detection of pancreatic cancer. MicroRNAs (miRs), a type of small non-coding RNA molecules, have emerged as highly sensitive molecular diagnostic tools that have the potential to function as precise indicators for a range of diseases, including cancer. Biosensors have been suggested as a potential solution for tackling these challenges, offering a promising approach for the early detection of pancreatic cancer. Small non-coding RNA molecules known as MicroRNAs (miRs) have become recognized as extremely sensitive molecular diagnostic tools and can act as precise biomarkers for different diseases, such as cancer. Moreover, this manuscript presents a thorough summary of the latest innovations in nano-biosensors that have been specifically developed for the identification of non-coding RNAs related to pancreatic cancer.
Collapse
Affiliation(s)
| | - Mahtab Hatami-Araghi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Majid Jafari-Khorchani
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Oushyani Roudsari
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Mona Abdolrahmat
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hassan Ghasemi
- Research Center for Environmental Contaminants (RCEC), Abadan University of Medical Sciences, Abadan, Iran.
| | - Saleh Aiiashi
- Abadan University of Medical Sciences, Abadan, Iran.
| |
Collapse
|
9
|
Jian Z, Pan T, Li R, Zhang W, Cheng T, Zhang H, Song J, Shi N, Zhang Z. Comprehensive analysis of UPK3B as a marker for prognosis and immunity in pancreatic adenocarcinoma. Sci Rep 2025; 15:12716. [PMID: 40223017 PMCID: PMC11994762 DOI: 10.1038/s41598-025-97213-x] [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: 10/09/2024] [Accepted: 04/03/2025] [Indexed: 04/15/2025] Open
Abstract
The low immunogenicity of pancreatic cancer inhibits effective antitumor immune responses, primarily due to the immune evasion mediated by low expression of the major histocompatibility complex (MHC). Through comprehensive analysis, our study identifies UPK3B as a gene closely associated with low MHC expression and low immunogenicity in pancreatic cancer. UPK3B has been reported as a marker of primary mesothelial cells, mature epicardium and promotes extracellular matrix signaling. However, the role of UPK3B in pancreatic cancer remain unclear. We found that UPK3B is highly predictive of overall survival (OS) in patients with pancreatic ductal adenocarcinoma (PDAC) and is significantly related to clinical features, immune cell infiltration, and response to immune checkpoint inhibitor (ICI) therapy. Gene enrichment analysis revealed significant downregulation of immune regulatory and BCR signaling pathways in the UPK3B high-expression group. Additionally, UPK3B is positively correlated with immunosuppressive cells, suggesting that high UPK3B expression may inhibit antitumor immune responses by promoting low MHC expression. UPK3B is also positively correlated with immune checkpoints, indicating that tumors with high UPK3B expression may not benefit from ICI therapy. Therefore, UPK3B may serve as a novel biomarker and therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Ziying Jian
- Department of Hematology, Zhong da Hospital of Southeast University, Nanjing, China
| | - Tao Pan
- Department of Radiology, Center of Interventional Radiology and Vascular Surgery, Medical School, Zhongda Hospital, Southeast University, Nanjing, China
| | - Renjie Li
- School of Medicine, Southeast University, Nanjing, China
| | - Weiyu Zhang
- Department of General Surgery, Zhongda Hospital of Southeast University, Nanjing, China
| | - Tao Cheng
- Department of General Surgery, Zhongda Hospital of Southeast University, Nanjing, China
| | - Hanzhe Zhang
- School of Medicine, Southeast University, Nanjing, China
| | - Jialin Song
- School of Medicine, Southeast University, Nanjing, China
| | - Naipeng Shi
- Department of Urology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Zhiheng Zhang
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
10
|
Sharma R, Kumar S, Komal K, Ghosh R, Thakur S, Pal RR, Kumar M. Comprehensive insights into pancreatic cancer treatment approaches and cutting-edge nanocarrier solutions: from pathology to nanomedicine. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04094-y. [PMID: 40202672 DOI: 10.1007/s00210-025-04094-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025]
Abstract
Pancreatic cancer is one of the most lethal malignancies worldwide. It is characterized by poor prognosis, high mortality, and recurrence rates. Various modifiable and non-modifiable risk factors are associated with pancreatic cancer incidence. Available treatments for pancreatic cancer include surgery, chemotherapy, radiotherapy, photodynamic therapy, supportive care, targeted therapy, and immunotherapy. However, the survival rates for PC are very low. Regrettably, despite efforts to enhance prognosis, the survival rate of pancreatic cancer remains relatively low. Therefore, it is essential to investigate new approaches to improve pancreatic cancer treatment. By synthesizing current knowledge and identifying existing gaps, this article provides a comprehensive overview of risk factors, pathology, conventional treatments, targeted therapies, and recent advancements in nanocarriers for its treatment, along with various clinical trials and patents that justify the safety and efficacy of innovative carriers for drug delivery systems. Ultimately, this review underscores the potential of these innovative formulations to improve outcomes and contribute significantly to the advancement of Pancreatic Cancer treatment. Together, these insights highlight nano-formulations as a promising frontier for effectively treating Pancreatic Cancer.
Collapse
Affiliation(s)
- Rohit Sharma
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Kumari Komal
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Rashmi Ghosh
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Shubham Thakur
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Ravi Raj Pal
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
11
|
Nakashima M, Fukumoto A, Matsuda S. Beneficial Probiotics with New Cancer Therapies for Improved Treatment of Hepatocellular Carcinoma. Diseases 2025; 13:111. [PMID: 40277821 DOI: 10.3390/diseases13040111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant form of primary liver cancer. Intricate networks linked to the host immune system may be associated with the pathogenesis of HCC. A huge amount of interdisciplinary medical information for the treatment of HCC has been accumulated over recent years. For example, advances in new immunotherapy have improved the results of treatment for HCC. This approach can be advantageously combined with standard conventional treatments such as surgical resection to improve the therapeutic effect. However, several toxic effects of treatments may pose a significant threat to human health. Now, a shift in mindset is important for achieving superior cancer therapy, where probiotic therapy may be considered, at least within the bounds of safety. The interplay between the gut microbiota and immune system could affect the efficacy of several anticancer treatments, including of immune checkpoint therapy via the alteration of Th17 cell function against various malignant tumors. Here, some recent anticancer techniques are discussed, whereby the growth of HCC may be effectively and safely repressed by probiotic therapy.
Collapse
Affiliation(s)
- Moeka Nakashima
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Akari Fukumoto
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
12
|
Okasha HH, Tag-Adeen M, Shaaban HE. Role of pancreatic juice cytology in diagnosis of high-grade pancreatic intraepithelial neoplasia. World J Clin Cases 2025; 13:94437. [PMID: 40191674 PMCID: PMC11670036 DOI: 10.12998/wjcc.v13.i10.94437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 11/05/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024] Open
Abstract
High-grade pancreatic intraepithelial neoplasia is a challenging diagnosis and it does not exhibit mass lesions. It is suspected based on changes in the main pancreatic duct in magnetic resonance cholangiopancreatography. Sometimes only an unclear duct shows in magnetic resonance cholangiopancreatography with no focal strictures and upstream dilatation of the main pancreatic duct. Serial pancreatic juice cytology is valuable in diagnosis of those patients.
Collapse
Affiliation(s)
- Hussein Hassan Okasha
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kasr Al-Aini School of Medicine, Cairo University, Cairo 11562, Egypt
| | - Mohammed Tag-Adeen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Qena Faculty of Medicine, South Valley University, Qena 83523, Egypt
| | - Hossam Eldin Shaaban
- Department of Internal Medicine and Gastroenterology, National Hepatology and Tropical Medicine Research Institute, Cairo 11796, Egypt
| |
Collapse
|
13
|
Wang LH, Jiang Y, Sun CH, Chen PT, Ding YN. Advancements in the application of ablative therapy and its combination with immunotherapy in anti-cancer therapy. Biochim Biophys Acta Rev Cancer 2025; 1880:189285. [PMID: 39938664 DOI: 10.1016/j.bbcan.2025.189285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 02/14/2025]
Abstract
Cancer is a significant health issue impacting humans. Currently, systemic therapies such as chemotherapy have significantly increased the life expectancy of cancer patients. However, some patients are unable to endure systemic treatment due to its significant adverse effects, leading to an increased focus on local therapies including radiation and ablation therapy. Ablation therapy is a precise, low-toxicity, and minimally invasive localized therapy that is increasingly acknowledged by clinicians and cancer patients. Many cancer patients have benefited from it, with some achieving full recovery. Currently, numerous studies have shown that ablation therapy is effective due to its ability to kill cancer cells efficiently and activate the body's anti-cancer immunity. It can also convert "cold cancers" into "hot cancers" and enhance the effectiveness of immunotherapy when used in combination. In this article, we categorize ablation therapy into thermal ablation, cryoablation, photodynamic therapy (PDT), irreversible electroporation (IRE), etc. Thermal ablation is further divided into Radiofrequency ablation (RFA), microwave ablation (WMA), high-frequency focused ultrasound (HIFU), photothermal therapy (PTT), magnetic heat therapy (MHT), etc. We systematically review the most recent advancements in these ablation therapies that are either currently used in clinic or are anticipated to be used in clinic. Then, we also review the latest development of various ablative therapies combined with immunotherapy, and its future development. CLINICAL RELEVANCE STATEMENT: Ablation therapy, an invasive localized treatment, offers an alternative to systemic therapies for cancer patients who cannot tolerate their adverse effects. Its ability to kill cancer cells efficiently and activate anti-cancer immunity. This article reviews recent advancements in ablation therapies, including thermal, cryoablation, PDT, and IRE, and their potential clinical applications, both standalone and in combination with immunotherapy.
Collapse
Affiliation(s)
- Lu-Hong Wang
- Department of Interventional Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Center of Interventional Radiology & Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, China; State Key Laboratory of Digital Medical Engineering, National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | - Yi Jiang
- Department of Interventional Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Hangzhou, Zhejiang 310022, China; Zhejiang Provincial Research Center for Innovative Technology and Equipment in Interventional Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Chen-Hang Sun
- Department of Interventional Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Hangzhou, Zhejiang 310022, China; Zhejiang Provincial Research Center for Innovative Technology and Equipment in Interventional Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Peng-Tao Chen
- Department of Interventional Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Hangzhou, Zhejiang 310022, China; Zhejiang Provincial Research Center for Innovative Technology and Equipment in Interventional Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Yi-Nan Ding
- Department of Interventional Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Hangzhou, Zhejiang 310022, China; Zhejiang Provincial Research Center for Innovative Technology and Equipment in Interventional Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| |
Collapse
|
14
|
Powrózek T, Otieno MO, Maffeo D, Frullanti E, Martinez-Useros J. Blood circulating miRNAs as pancreatic cancer biomarkers: An evidence from pooled analysis and bioinformatics study. Int J Biol Macromol 2025:142469. [PMID: 40180095 DOI: 10.1016/j.ijbiomac.2025.142469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 03/09/2025] [Accepted: 03/22/2025] [Indexed: 04/05/2025]
Abstract
Pancreatic cancer (PC) is one of the deadliest cancers, characterized by a poor prognosis. Currently, there are no screening programs for the early detection of PC, and existing diagnostic methods are primarily limited to high-risk individuals. Biomarkers such as CA19-9 have not significantly improved early diagnosis, making the identification of new potential biomarkers crucial for routine clinical practice. Among the candidate biomarkers, miRNAs have been most extensively studied due to their role in regulating gene expression (either as oncomiRs or tumor suppressor miRNAs) and their potential for minimally invasive analysis through liquid biopsy techniques. This review aims to summarize the current literature on blood-circulating miRNAs and their diagnostic value in PC detection, considering the context of CA19-9 and benign pancreatic diseases. The data from the collected studies were curated through both statistical and bioinformatics analyses to identify the most promising miRNAs with optimal diagnostic accuracy for PC detection and to assess their role in the molecular processes leading to tumor development.
Collapse
Affiliation(s)
- Tomasz Powrózek
- Department of Human Physiology, Medical University of Lublin, Lublin, Poland.
| | - Michael Ochieng' Otieno
- Translational Oncology Division, Oncohealth Institute, Fundacion Jiménez Díaz University Hospital, Madrid, Spain
| | - Debora Maffeo
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Cancer Genomics and Systems Biology Lab, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elisa Frullanti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Cancer Genomics and Systems Biology Lab, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Javier Martinez-Useros
- Translational Oncology Division, Oncohealth Institute, Fundacion Jiménez Díaz University Hospital, Madrid, Spain; Area of Physiology, Department of Basic Health Sciences, Faculty of Health Sciences, Rey Juan Carlos University, Madrid, Spain
| |
Collapse
|
15
|
Ding Y, Yu Y. Therapeutic potential of flavonoids in gastrointestinal cancer: Focus on signaling pathways and improvement strategies (Review). Mol Med Rep 2025; 31:109. [PMID: 40017144 PMCID: PMC11884236 DOI: 10.3892/mmr.2025.13474] [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: 09/18/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
Flavonoids are a group of polyphenolic compounds distributed in vegetables, fruits and other plants, which have considerable antioxidant, anti‑tumor and anti‑inflammatory activities. Several types of gastrointestinal (GI) cancer are the most common malignant tumors in the world. A large number of studies have shown that flavonoids have inhibitory effects on cancer, and they are recognized as a class of potential anti‑tumor drugs. Therefore, the present review investigated the molecular mechanisms of flavonoids in the treatment of different types of GI cancer and summarized the drug delivery systems commonly used to improve their bioavailability. First, the classification of flavonoids and the therapeutic effects of various flavonoids on human diseases were briefly introduced. Then, to clarify the mechanism of action of flavonoids on different types of GI cancer in the human body, the metabolic process of flavonoids in the human body and the associated signaling pathways causing five common types of GI cancer were discussed, as well as the corresponding therapeutic targets of flavonoids. Finally, in clinical settings, flavonoids have poor water solubility, low permeability and inferior stability, which lead to low absorption efficiency in vivo. Therefore, the three most widely used drug delivery systems were summarized. Suggestions for improving the bioavailability of flavonoids and the focus of the next stage of research were also put forward.
Collapse
Affiliation(s)
- Ye Ding
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yong Yu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
16
|
Rostom MM, Rashwan AA, Sotiropoulou CD, Hozayen SZ, Abdelhamid AM, Abdelhalim MM, Eltahtawy O, Emara HM, Elemam NM, Kontos CK, Youness RA. MIAT: A pivotal oncogenic long noncoding RNA tunning the hallmarks of solid malignancies. Transl Oncol 2025; 54:102329. [PMID: 40014977 PMCID: PMC11910686 DOI: 10.1016/j.tranon.2025.102329] [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: 10/22/2024] [Revised: 01/06/2025] [Accepted: 02/13/2025] [Indexed: 03/01/2025] Open
Abstract
Long non-coding RNAs (LncRNAs) have emerged as intriguing players in cellular regulation, challenging the traditional view of non-coding RNAs as mere "dark genome". Non-coding DNA makes up most of the human genome and plays a pivotal role in cancer development. These RNA molecules, which do not code for proteins, have captivated researchers with their diverse and crucial roles in gene regulation, chromatin dynamics, and other cellular processes. In several physiological and pathological circumstances, lncRNAs serve critical functions. This review will tackle the complex function of the lncRNA myocardial infarction-associated transcript (MIAT) in various solid malignancies. A special emphasis would be directed on the correlation between cancer patients' clinicopathological features and the expression profile of MIAT. MIAT is a oncogenic regulator in many malignant tumors, where it can control the growth, invasion, metastasis, and resistance to death of cells. As a result, MIAT is thought to be a possible biomarker and therapeutic target for cancer patients. The biological functions, mechanisms and potential clinical implications of MIAT during carcinogenesis and finally the current possible therapeutic approaches targeting MIAT are also outlined in this review.
Collapse
Affiliation(s)
- Monica M Rostom
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835, Cairo, Egypt
| | - Alaa A Rashwan
- Biotechnology Graduate Program, School of Sciences and Engineering, The American University in Cairo (AUC), 11835, Cairo, Egypt
| | - Christina D Sotiropoulou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701, Athens, Greece
| | - Sama Z Hozayen
- Molecular Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU), 11835, Cairo, Egypt
| | | | - Miriam Mokhtar Abdelhalim
- Molecular Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU), 11835, Cairo, Egypt
| | - Omar Eltahtawy
- Molecular Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU), 11835, Cairo, Egypt
| | - Hadir M Emara
- Molecular Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU), 11835, Cairo, Egypt; Department of Nanotechnology, School of Sciences and Engineering, The American University in Cairo (AUC), 11835, Cairo, Egypt
| | - Noha M Elemam
- Clinical Sciences Department, College of Medicine, University of Sharjah, 27272, Sharjah, UAE; Research Institute for Medical and Health Sciences, University of Sharjah, 27272, Sharjah, UAE
| | - Christos K Kontos
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701, Athens, Greece
| | - Rana A Youness
- Molecular Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU), 11835, Cairo, Egypt.
| |
Collapse
|
17
|
Tabacelia D, Robles-Medranda C, Klimko A, Pereira SP, Vilmann P, Voermans RP, Săftoiu A, Tieranu CG, Stroescu C. Technical Feasibility of Microwave Ablation in Pancreatic Tumors: A Scoping Review of Procedural Efficacy and Safety. Cancers (Basel) 2025; 17:1197. [PMID: 40227787 PMCID: PMC11988054 DOI: 10.3390/cancers17071197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pancreatic cancer remains one of the most aggressive and lethal malignancies, with limited effective treatment options for advanced stages. Microwave Ablation (MWA) has emerged as a minimally invasive therapeutic modality, offering potential benefits in tumor control. This review aims to critically assess the safety and efficacy of MWA in the treatment of pancreatic cancer, focusing on its application in various pancreatic lesions. METHODS We systematically reviewed studies published between 2010 and 2023 that evaluated the use of MWA in pancreatic tumors, including locally advanced pancreatic cancer (LAPC), pancreatic neuroendocrine tumors (PNETs), and pancreatic metastases from renal cell carcinoma (RCC). Due to limited data on survival rates and long-term outcomes, our analysis concentrated primarily on the technical aspects and immediate procedural outcomes of MWA. RESULTS MWA was technically feasible in all cases. The overall complication rate was approximately 16.7% (nine patients), with higher incidences in tumors located in the pancreatic head. Reported complications included pancreatitis and pseudocyst formation. Procedural parameters varied, with applied energy ranging from 20 to 80 watts and ablation times between 2 to 15 min, depending on the microwave generator type and approach (percutaneous, intraoperative or endoscopic). All cases demonstrated effective necrosis of the target tissue, and several studies reported notable tumor size reductions, averaging up to 70%. CONCLUSIONS MWA shows promise as a therapeutic option for pancreatic cancer, achieving high technical success rates and significant tumor reductions. However, the procedure is associated with a moderate complication rate, particularly in tumors located in the pancreatic head.
Collapse
Affiliation(s)
- Daniela Tabacelia
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 010017 Bucharest, Romania; (D.T.); (A.S.); (C.G.T.)
- Elias Emergency University Hospital, 010017, Bucharest, Romania
| | | | | | - Stephen P. Pereira
- Institute for Liver and Digestive Health, University College London, London NW3 2QG, UK;
| | - Peter Vilmann
- Gastro Unit, Division of Endoscopy, Herlev and Gentofte Hospital, 2730 Herlev, Denmark;
- Department of Clinical Medicine, University of Copenhagen, 1050 Copenhagen, Denmark
| | - Rogier P. Voermans
- Department of Gastroenterology & Hepatology, Amsterdam Gastroenterology and Metabolism, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
| | - Adrian Săftoiu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 010017 Bucharest, Romania; (D.T.); (A.S.); (C.G.T.)
- Elias Emergency University Hospital, 010017, Bucharest, Romania
| | - Cristian George Tieranu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 010017 Bucharest, Romania; (D.T.); (A.S.); (C.G.T.)
- Elias Emergency University Hospital, 010017, Bucharest, Romania
| | - Cezar Stroescu
- Surgery Department, Fundeni Clinical Institute, 010017 Bucharest, Romania;
| |
Collapse
|
18
|
Sulekha Suresh D, Jain T, Dudeja V, Iyer S, Dudeja V. From Microbiome to Malignancy: Unveiling the Gut Microbiome Dynamics in Pancreatic Carcinogenesis. Int J Mol Sci 2025; 26:3112. [PMID: 40243755 PMCID: PMC11988718 DOI: 10.3390/ijms26073112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/01/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
Pancreatic cancer is a major cause of cancer-associated mortality globally, characterized by a poor prognosis and limited therapeutic response. The current approach for treating pancreatic cancer involves locoregional control with surgical resection and systemic therapy in the form of cytotoxic chemotherapy. However, despite standard-of-care treatment, the outcomes remain dismal. Emerging evidence suggests that the gut microbiota plays a significant role in pancreatic carcinogenesis through dysbiosis, chronic inflammation and immune modulation. Dysbiosis-driven alterations in the gut microbiota composition can disrupt intestinal homeostasis, promote systemic inflammation and create a tumor-permissive microenvironment in the pancreas. Moreover, the gut microbiota modulates the efficacy of systemic therapies, including chemotherapy and immunotherapy, by impacting drug metabolism and shaping the tumor immune landscape. This review is mainly focused on exploring the intricate interplay between the gut microbiota and pancreatic cancer, and also highlighting its dual role in carcinogenesis and the therapeutic response.
Collapse
Affiliation(s)
| | | | | | | | - Vikas Dudeja
- Division of Surgical Oncology, Department of Surgery, The University of Alabama at Birmingham, BDB 573 1808 7th Avenue South, Birmingham, AL 35294, USA; (D.S.S.); (T.J.); (V.D.); (S.I.)
| |
Collapse
|
19
|
Zhang J, Sun W, Wu W, Qin Z, Wei B, Li T. METTL3-dependent m6A methylation of circCEACAM5 fuels pancreatic cancer progression through DKC1 activation. Cell Mol Life Sci 2025; 82:132. [PMID: 40146281 PMCID: PMC11950576 DOI: 10.1007/s00018-025-05653-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/20/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Pancreatic cancer is highly lethal and has a poor prognosis. Research has highlighted the role of circular RNAs and m6A methylation in cancer progression. METTL3, a key m6A methyltransferase, is linked to various cancers, but its interaction with circular RNAs in pancreatic cancer is unclear. This study examined the role of circCEACAM5 in pancreatic cancer, particularly its regulation by METTL3-mediated m6A methylation and interaction with effectors such as DKC1. METHODS circCEACAM5 expression in pancreatic cancer tissues and cell lines was evaluated via RT‒qPCR. Its characteristics were validated through Sanger sequencing, stability assays, and FISH. Functional assays (CCK-8, EdU, Transwell, and flow cytometry) were conducted in AsPC-1 cells, and in vivo tumor models were established. m6A modification was analyzed via bioinformatics tools and m6A-specific immunoprecipitation, while RNA pull-down assays were used to examine the interaction of circCEACAM5 with METTL3 and DKC1. RESULTS circCEACAM5 was significantly upregulated in pancreatic cancer and correlated with poor clinical outcomes. CircCEACAM5 promoted cell proliferation, invasion, and migration while inhibiting apoptosis both in vitro and in vivo. METTL3-mediated m6A methylation of circCEACAM5 was confirmed, and METTL3 knockdown reversed the effects of circCEACAM5 silencing on the malignant behavior of pancreatic cancer cells. circCEACAM5 interacted with DKC1, and DKC1 overexpression reversed the effects of circCEACAM5 knockdown on the malignant behavior of pancreatic cancer cells. CONCLUSION METTL3-mediated m6A methylation of circCEACAM5 drives pancreatic cancer progression by increasing DKC1 expression, suggesting potential new therapeutic targets for this aggressive malignancy.
Collapse
Affiliation(s)
- Jie Zhang
- School of Biology and Food Engineering, Changshu Institute of Technology, Suzhou, 215500, People's Republic of China
| | - Wenxue Sun
- Jining First People's Hospital, Jining Medical University, Jining, 272000, People's Republic of China
| | - Wenda Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Zihui Qin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Ben Wei
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Tushuai Li
- School of Biology and Food Engineering, Changshu Institute of Technology, Suzhou, 215500, People's Republic of China.
| |
Collapse
|
20
|
Zheng BW, Yang XY, Zheng J, Yao RC. Dynamic evaluation of postoperative survival in pancreatic ductal adenocarcinoma. Medicine (Baltimore) 2025; 104:e41942. [PMID: 40128033 PMCID: PMC11936588 DOI: 10.1097/md.0000000000041942] [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: 11/27/2024] [Accepted: 03/06/2025] [Indexed: 03/26/2025] Open
Abstract
The pancreatic ductal adenocarcinoma has a high degree of malignancy, and traditional prognostic assessment methods have limited evaluative capacity. This study is based on the Kallikrein-related peptidase 7 (KLK7) expression and uses conditional survival algorithms to perform dynamic survival assessments of patients. The Cox proportional hazards model was employed for identifying and adjusting for potential confounders. The Kaplan-Meier technique was utilized to estimate the overall survival rate. The computation of the likelihood of patients surviving an additional year after X years of survival was achieved using the equation CS1 = OS(X + 1)/OS(X). A subgroup analysis based on CS1 was conducted for each individual risk factor. A total of 243 eligible patients were included in the study. Conditioned survival (CS) refers to the years a patient has already survived and the predicted years they are likely to survive in the future, while conducting a time-varying analysis of the factors influencing prognosis. The survival probability assessed by CS1 increased year by year, with the 1-, 2-, and 3-year survival rates rising from 50.4% to 91.2%. In contrast, the actuarial overall survival (OS) decreased from 81.9% at 1 year to 38.6% at 3 years post-surgery. The results of the conditional analysis indicate that patients who survive longer within a certain timeframe have better survival expectations in the future. Adverse factors, including KLK7, have a decreasing impact on survival over time. Conditional survival analysis based on KLK7 can provide more accurate survival predictions for patients who has identified KLK7.
Collapse
Affiliation(s)
- Bo-Wen Zheng
- Department of Hepato-Pancreato-Biliary Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
- Yichang Central People’s Hospital, Yichang, Hubei, China
| | - Xin-Yu Yang
- Department of Hepato-Pancreato-Biliary Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
- Yichang Central People’s Hospital, Yichang, Hubei, China
| | - Jun Zheng
- Department of Hepato-Pancreato-Biliary Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
- Yichang Central People’s Hospital, Yichang, Hubei, China
| | - Ru-Cheng Yao
- Department of Hepato-Pancreato-Biliary Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
- Yichang Central People’s Hospital, Yichang, Hubei, China
| |
Collapse
|
21
|
Kuchenreuther I, Clausen FN, Mazurie J, Paul S, Czubayko F, Mittelstädt A, Koch AK, Karabiber A, Hansen FJ, Arnold LS, Weisel N, Merkel S, Brunner M, Krautz C, Vera J, Grützmann R, Weber GF, David P. Increased Herpesvirus Entry Mediator Expression on Circulating Monocytes and Subsets Predicts Poor Outcomes in Pancreatic Ductal Adenocarcinoma Patients. Int J Mol Sci 2025; 26:2875. [PMID: 40243455 PMCID: PMC11988668 DOI: 10.3390/ijms26072875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is aggressive, with a 5-year survival rate of only 12.8%, and its increasing incidence in Western countries highlights the urgent need for better early-stage detection and treatment methods. Early diagnosis significantly improves the chances of survival, but non-specific symptoms and undetectable precursor lesions pose a major challenge. To date, there are no reliable screening tools to detect PDAC at an early stage. Herpesvirus entry mediator (HVEM) has already been proposed as a prognostic marker in numerous cancer types. Therefore, we investigated the role of HVEM in PDAC. Flow cytometry was used to analyze HVEM expression in immune cells and its inhibitory receptors (CD160 and BTLA) on T-cells, as well as its subsets in the peripheral blood of 57 diagnosed PDAC patients and 17 clinical controls. In addition, survival analyses were performed within the PDAC cohort, changes in HVEM expression were analyzed in relation to clinicopathological parameters, and a correlation analysis between HVEM expression and cytokine levels of IL-6 and IL-10 was conducted. Furthermore, HVEM expression on monocytes and their subsets was evaluated as a potential prognostic marker and compared with the prognostic utility of CA19-9. We found that HVEM expression is significantly elevated on immune cells, particularly on monocytes (p < 0.0001) and their subsets, in PDAC patients, and is associated with reduced survival (p = 0.0067) and clinicopathological features such as perineural, lymphovascular, and vascular invasion. Moreover, HVEM-expressing monocytes demonstrated superior predictive value compared to CA19-9, highlighting their potential as part of a combined screening tool for PDAC. In conclusion, HVEM on monocytes could serve as a novel prognostic marker for PDAC.
Collapse
MESH Headings
- Humans
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- Receptors, Tumor Necrosis Factor, Member 14/blood
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/blood
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/diagnosis
- Female
- Male
- Middle Aged
- Monocytes/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/blood
- Pancreatic Neoplasms/mortality
- Prognosis
- Aged
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/metabolism
- Antigens, CD/metabolism
- Receptors, Immunologic/metabolism
- Interleukin-10
- Adult
- Interleukin-6
- GPI-Linked Proteins
Collapse
Affiliation(s)
- Isabelle Kuchenreuther
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Finn-Niklas Clausen
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Johanne Mazurie
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Sushmita Paul
- Department of Dermatology, University Hospital Erlangen, 91054 Erlangen, Germany; (S.P.); (J.V.)
| | - Franziska Czubayko
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Anke Mittelstädt
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Ann-Kathrin Koch
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Alara Karabiber
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Frederik J. Hansen
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Lisa-Sophie Arnold
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Nadine Weisel
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Susanne Merkel
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Maximilian Brunner
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Christian Krautz
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Julio Vera
- Department of Dermatology, University Hospital Erlangen, 91054 Erlangen, Germany; (S.P.); (J.V.)
| | - Robert Grützmann
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| | - Georg F. Weber
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| | - Paul David
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
22
|
Simon M, Meurat A, Stanislas S, Dréau H, Belleannée G, Laurent C, Blanc JF, Mounicou S. Metallic trace elements in pancreatic tissue are associated with higher risk of pancreatic adenocarcinoma - METAPANDA study. Pancreatology 2025:S1424-3903(25)00059-6. [PMID: 40169314 DOI: 10.1016/j.pan.2025.03.006] [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: 12/24/2024] [Revised: 03/09/2025] [Accepted: 03/17/2025] [Indexed: 04/03/2025]
Abstract
OBJECTIVES Few recent studies have indicated the possible involvement of some metallic trace element (MTE), commonly known as heavy metals, in pancreatic ductal adenocarcinoma (PDAC). To evaluate the potential role of MTE in PDAC onset, we compared concentrations of 23 MTE in healthy pancreas tissue close to the tumor, from patients who underwent pancreatic surgery for PDAC in cases group and for neuroendocrine or other lesion for controls. METHODS Samples were taken from paraffin-embedded pancreatectomy blocks of 33 PDAC cases and 29 controls. Concentrations of 23 MTE were determined by inductively coupled plasma mass spectrometry (ICP-MS). RESULTS In multivariate analysis, associations were found between risk of PDAC and higher tissue concentration of antimony (OR 6.31, 95 % CI 2.06-29.03; p = 0.006), thallium (OR 3.23, 95 % CI 1.35-12.07; p = 0.033) arsenic (OR 2.96, 95 % CI 1.22-10.10; p = 0.04) and lead (OR 2.27, 95 % CI 1.13-5.77; p = 0.044). CONCLUSIONS This pilot study presents unpublished information about a large set of MTE in pancreatic tissues, confirming the possible involvement of arsenic and lead in PDAC onset and highlighting the potential role of antimony and thallium which have never been implicated before.
Collapse
Affiliation(s)
- Mireille Simon
- Pau Hospital Center, Department of Gastroenterology and Digestive Oncology, 64000, Pau, France.
| | - Aurore Meurat
- Bordeaux University Hospital, Department of Digestive Oncology, Haut Leveque Hospital, 33604, Pessac, France
| | - Sophie Stanislas
- Pau Hospital Center, Department of Pathology, 64000, Pau, France
| | - Hervé Dréau
- Pau Hospital Center, Department of Public Health and Social Medicine, 64000, Pau, France
| | - Geneviève Belleannée
- Bordeaux University Hospital, Department of Pathology, Haut Leveque Hospital, 33604, Pessac, France
| | - Christophe Laurent
- Bordeaux University Hospital, Department of Digestive Surgery, Haut Leveque Hospital, 33604, Pessac, France
| | - Jean-Frédéric Blanc
- Bordeaux University Hospital, Department of Digestive Oncology, Haut Leveque Hospital, 33604, Pessac, France
| | - Sandra Mounicou
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, 64000, Pau, France
| |
Collapse
|
23
|
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
|
24
|
Du Y, Zhao Y, Li J, Wang J, You S, Zhang Y, Zhang L, Yang J, Alinejad-Rokny H, Cheng S, Shao C, Zou D, Ye Y. PLXDC1 + Tumor-Associated Pancreatic Stellate Cells Promote Desmoplastic and Immunosuppressive Niche in Pancreatic Ductal Adenocarcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2415756. [PMID: 40091495 DOI: 10.1002/advs.202415756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/27/2025] [Indexed: 03/19/2025]
Abstract
Pancreatic stellate cells (PSCs) contribute to pancreatic ductal adenocarcinoma (PDAC) progression and therapeutic resistance, yet their detailed functions remain unclear. This study combined RNA sequencing and assay for transposase-accessible chromatin using sequencing (ATAC-seq) on sorted PSCs from adjacent normal and PDAC tissues to investigate their transcriptional and epigenetic activation. PSCs heterogeneity and functions are characterized through bulk, single-cell, and spatial transcriptomes, as well as in situ sequencing. The clinical relevance of PSCs in immunotherapy is assessed using an in-house immune-checkpoint blockade (ICB) treatment cohort. Findings showed that stress and hypoxia signaling activated PSCs in PDAC. Three common PSCs (CPSCs) and four tumor-associated PSCs (TPSCs) are identified, each with distinct functions. CPSCs differentiated into CCL19+ TPSCs in immune-enriched regions, MYH11+ TPSCs in the stromal region, and PLXDC1+ TPSCs, which exhibited cancer-associated myofibroblasts (myCAFs) phenotype linked to poor prognosis. Notably, PLXDC1+ TPSCs, located near aggressive LRRC15+ myCAFs and SPP1+ macrophages, formed a desmoplastic and immunosuppressive niche around the tumor boundary, promoting CD8 T cell exhaustion. Single-cell transcriptomics of PDAC patients treated with ICB revealed that PLXDC1+ TPSCs correlated with poor immunotherapy efficacy. Overall, this study provides key insights into PSCs in PDAC and potential therapeutic targets.
Collapse
Affiliation(s)
- Yanhua Du
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, China
| | - Yizhou Zhao
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Judong Li
- Department of Pancreatic-biliary Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Jiaxin Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Shenglan You
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Yao Zhang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Li Zhang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, China
| | - Jihong Yang
- Department of Hepatobiliary Surgery, Hebei Key Laboratory of General Surgery for Digital Medicine, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Hamid Alinejad-Rokny
- UNSW BioMedical Machine Learning Lab (BML), School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Shujie Cheng
- Department of Hepatobiliary Surgery, Hebei Key Laboratory of General Surgery for Digital Medicine, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Chenghao Shao
- Department of Pancreatic-biliary Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Duowu Zou
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Youqiong Ye
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, China
| |
Collapse
|
25
|
Myneni R, Pathak P, Hacker-Prietz A, He J, Kumar R, Narang AK. Effect of travel distance on utilization of ancillary services among patients with pancreatic ductal adenocarcinoma: A single institution study. Support Care Cancer 2025; 33:282. [PMID: 40088289 DOI: 10.1007/s00520-025-09339-3] [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: 09/24/2024] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Fragmentation of care among patients with pancreatic cancer between specialized tertiary centers and community centers may be associated with increased patient burden and poorer outcomes. However, the impact of distance from a tertiary center on utilization of key ancillary services such as dietician consultation, and palliative or pain medicine consultation is unclear. We sought to examine how this travel distance influences the utilization of key ancillary services. METHODS This retrospective cohort study included 200 consecutive patients who were seen for a diagnosis of pancreatic ductal adenocarcinoma (PDAC) in 2021. Patients were grouped by travel distance: < 12.5, 12.5-49, and > 50 miles. Demographics, disease staging, and use of key ancillary services such as, dietician consultation, palliative, and pain medicine consultation were compared. Multiple logistic regression assessed associations between travel distance and ancillary service utilization. RESULTS Of the 200 patients, 14.5% traveled < 12.5 miles, 39.5% traveled 12.5-49 miles, and 46% traveled over 50 miles to our institution. Patients living over 50 miles away were significantly more likely to receive chemotherapy and radiation locally (81.8% vs 44.4%, p < 0.001). Importantly, they were less likely to utilize key ancillary services, including registered dietician consultation (Odds Ratio (OR) 0.34, p = 0.03), pancreatic enzyme prescriptions (OR 0.35, p = 0.03), pain medicine consultation (OR 0.20, p < 0.01), and palliative care consultation (OR 0.24, p < 0.01) compared to those living closer. CONCLUSIONS Patients living over 50 miles from our institution were significantly less likely to receive key supportive services. Despite similar clinical characteristics, these disparities show how initiatives are necessary to guarantee equitable access to comprehensive cancer care, regardless of geographic location.
Collapse
Affiliation(s)
- Revathi Myneni
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, 401 North Broadway/Suite 1440, Baltimore, MD, 21287, USA
- Department of Medical Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Surgical Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Priya Pathak
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, 401 North Broadway/Suite 1440, Baltimore, MD, 21287, USA
- Department of Medical Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Surgical Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Amy Hacker-Prietz
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, 401 North Broadway/Suite 1440, Baltimore, MD, 21287, USA
- Department of Medical Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Surgical Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jin He
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, 401 North Broadway/Suite 1440, Baltimore, MD, 21287, USA
- Department of Medical Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Surgical Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Rachit Kumar
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, 401 North Broadway/Suite 1440, Baltimore, MD, 21287, USA
- Department of Medical Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Surgical Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Amol K Narang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, 401 North Broadway/Suite 1440, Baltimore, MD, 21287, USA.
- Department of Medical Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Surgical Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Radiology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
26
|
Salahi‐Niri A, Zarand P, Mansouri N, Rastgou P, Yazdani O, Esbati R, Shojaeian F, Jahanbin B, Mohsenifar Z, Aghdaei HA, Ardalan FA, Safavi‐Naini SAA. Potential of Proliferative Markers in Pancreatic Cancer Management: A Systematic Review. Health Sci Rep 2025; 8:e70412. [PMID: 40051490 PMCID: PMC11882395 DOI: 10.1002/hsr2.70412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/12/2024] [Accepted: 01/13/2025] [Indexed: 03/09/2025] Open
Abstract
Background and Aims Pancreatic cancer is an aggressive malignancy with poor prognosis and limited treatment options. Chemotherapy remains a primary therapeutic approach, but patient responses vary significantly, emphasizing the need for reliable biomarkers. This review explores the potential role of proliferative markers, including Ki-67, PCNA, Cyclin D1, and PHH3, as predictive and prognostic indicators in pancreatic cancer management, aiming to enhance personalized treatment strategies. Methods We conducted a narrative review by searching Scopus, PubMed, and Google Scholar for studies focusing on Ki-67, PCNA, Cyclin D1, and PHH3 in relation to pancreatic cancer and chemotherapy. The literature was reviewed to evaluate the role of these markers in predicting chemotherapy response, tumor progression, and overall patient survival. Results The review highlights the clinical significance of these markers. Ki-67 and PCNA are associated with cell proliferation, while Cyclin D1 regulates cell cycle progression and PHH3 is linked to mitotic activity. High expression levels of these markers often correlate with increased tumor aggressiveness and poorer patient outcomes. Moreover, they show promise in predicting chemotherapy response, which can inform tailored therapeutic strategies. However, challenges remain, including standardization of detection methods and determination of optimal cutoff values. Conclusion Proliferative markers such as Ki-67, PCNA, Cyclin D1, and PHH3 hold potential as predictive and prognostic tools in pancreatic cancer management. Their integration into clinical practice could improve the accuracy of treatment decisions and enhance patient outcomes. Further research and validation are necessary to overcome existing challenges and optimize their application in personalized oncology.
Collapse
Affiliation(s)
- Aryan Salahi‐Niri
- Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Paniz Zarand
- Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Negar Mansouri
- Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Parvaneh Rastgou
- School of MedicineTabriz University of Medical SciencesTabrizIran
| | - Omid Yazdani
- Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Romina Esbati
- Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Fatemeh Shojaeian
- Sidney Kimmel Comprehensive Cancer Research CenterJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Behnaz Jahanbin
- Cancer Institute, Pathology Department, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | - Zhaleh Mohsenifar
- Department of Pathology, Ayatollah Taleghani Educational Hospital, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Hamid Asadzadeh Aghdaei
- Research, Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Farid Azmoudeh Ardalan
- Pathology and Laboratory Medicine Department, Imam Khomeini Hospital ComplexTehran University of Medical SciencesTehranIran
| | | |
Collapse
|
27
|
Prabhakaran P, Palanisamy CP, Shanmugam A, Damodharan P, Swaminathan H, Devaraj D, Ahalliya RM, Velliyur Kanniappan G. Isolation, structural characterization, and molecular docking studies on the bioactive compound from n-Hexane extract of Emilia sonchifolia (L.) DC against the pancreatic cancer target Aurora 2 Kinase. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2025; 22:165-172. [PMID: 39686876 DOI: 10.1515/jcim-2024-0290] [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: 08/26/2024] [Accepted: 09/23/2024] [Indexed: 12/18/2024]
Abstract
OBJECTIVES Natural flora historically has played a substantial part in drug development since they serve as active ingredients in medications and templates for the synthesis of novel pharmaceuticals. Emilia sonchifolia is a conventionally utilised therapeutic flora in Indian pharmacopoeia. Therefore, the current study is intended to separate, structurally describe and analyse the anti-pancreatic cancer potential of isolated natural bio-constituents from E. sonchifolia (L.) DC. METHODS n-Hexane extract using chromatographic techniques and structurally characterize them using spectroscopic techniques. Further, in silico molecular docking method was employed to investigate the bioactivity of the isolated compound against Aurora 2 Kinase (pancreatic cancer target protein). RESULTS A mixture of stigmasterol with straight-chain monounsaturated alcohol (C49H86O) was isolated and identified from the aerial parts of E. sonchifolia using chromatographic techniques. The docking results showed that the isolated natural compound of stigmasterol with straight-chain monounsaturated alcohol has good docking results compared with Food and Drug Administration-permitted medicine. CONCLUSIONS Based on the outcomes, it can be concluded that the stigmasterol with straight-chain monounsaturated alcohol may act as a novel inhibitor for Aurora 2 Kinase. In the future, it may lead to the development of drugs targeting Aurora 2 Kinase for the effective treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Pratibha Prabhakaran
- Center for Wireless Networks Applications (WNA), Amrita Vishwa Vidyapeetham, Kollam, India
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | | | - Abirami Shanmugam
- Department of Anaesthesiology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, India
| | - Prabhu Damodharan
- Centre for Bioinformatics, Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Harshini Swaminathan
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Dhivya Devaraj
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | | | - Gopalakrishnan Velliyur Kanniappan
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
- Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, India
| |
Collapse
|
28
|
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
|
29
|
Lee D, Liew MS, Fourlanos S, Choi J. Metformin use and pancreatic ductal adenocarcinoma outcomes: a narrative review. ANZ J Surg 2025; 95:313-320. [PMID: 39840695 DOI: 10.1111/ans.19405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/04/2025] [Accepted: 01/09/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Metformin is a diabetes medication with anti-mitotic properties. A narrative review was performed to investigate people using metformin and the risk of developing pancreatic ductal adenocarcinoma (PDAC) as well as survival outcomes in established PDAC. METHODS Relevant studies on metformin use and PDAC were retrieved from PubMed including observational studies on metformin and the risk of developing PDAC and survival outcomes in PDAC, and randomized controlled trials of metformin as a treatment in PDAC. RESULTS Of the 367 studies searched, 26 studies fulfilled the criteria for this review. Metformin was not consistently associated with a reduced risk of developing PDAC. However, metformin use, especially higher cumulative doses, in some studies was associated with longer survival in patients with established PDAC, especially in the subgroup with resectable PDAC. Metformin use was not associated with longer survival in more advanced (non-resectable metastatic) PDAC. CONCLUSION Metformin was not consistently associated with a reduced risk of developing PDAC. Metformin may be associated with overall survival benefits in patients with PDAC including the resectable PDAC subgroup but not in the metastatic PDAC subgroup. The evidence to date does not support the routine use of metformin as an adjuvant therapy for advanced PDAC.
Collapse
Affiliation(s)
- Dooyeon Lee
- Department of Surgery, Western Health, St. Albans, Victoria, Australia
| | - Mun Sem Liew
- Victorian Oncology Care, St John of God Specialist Centre, Berwick, Victoria, Australia
| | - Spiros Fourlanos
- Department of Diabetes & Endocrinology, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Julian Choi
- Department of Surgery, Western Health, St. Albans, Victoria, Australia
- Department of Surgery, University of Melbourne, Parkville, Victoria, Australia
- Clinical Institute General Surgery and Gastroenterology, Epworth Healthcare, Richmond, Victoria, Australia
| |
Collapse
|
30
|
Xie L, Gong J, He Z, Zhang W, Wang H, Wu S, Wang X, Sun P, Cai L, Wu Z, Wang H. A Copper-Manganese Based Nanocomposite Induces Cuproptosis and Potentiates Anti-Tumor Immune Responses. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412174. [PMID: 39955646 DOI: 10.1002/smll.202412174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/26/2025] [Indexed: 02/17/2025]
Abstract
Cancer is one of the most important challenges worldwide with an increasing incidence. However, most of patients with malignant cancer receiving traditional therapies have tumor recurrence and short-term 5-year survival. Herein, a novel Cu2O-MnO@PEG (CMP) nanomaterial is developed to treat tumors. CMP directly mediates cuproptosis in tumor cells. Meanwhile, CMP potentiates anti-tumor immune responses in the tumor microenvironment (TME) to induce tumor regression. CMP improves the tumor antigen processing and presentation of dendritic cells and tumor-associated macrophages, and further promotes CD8+ T cell responses, especially for cytotoxic CD8+ T cells and transitory exhausted CD8+ T cells. Additionally, CMP downregulates the proportion of Treg cells and CTLA-4 expression on Treg cells. Notably, CMP induces systemic immune responses against distant tumors and long-term immune memory. Furthermore, CMP synergized with PD-L1 mAb promotes tumor inhibition and sustains the anti-tumor efficacy post PD-L1 mAb treatment. Collectively, this strategy has the clinically therapeutic potential for tumors by facilitating cuproptosis in tumor cells and anti-tumor immune responses.
Collapse
Affiliation(s)
- Luoyingzi Xie
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Jie Gong
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
- School of Clinical Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
- Department of Hepatobiliary Surgery, Leshan People's Hospital, Leshan, 614000, P. R. China
| | - Zhiqiang He
- Department of Dermatology, Southwest Hospital Jiangbei Area (The 958th hospital of Chinese People's Liberation Army), Chongqing, 400020, P. R. China
| | - Weinan Zhang
- Department of Urinary Nephropathy Center, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, P. R. China
| | - Haoyu Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
| | - Shitao Wu
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
- Graduate School of Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Xianxing Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
| | - Pijiang Sun
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
| | - Lei Cai
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
| | - Zhongjun Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
| |
Collapse
|
31
|
Pitman MB. The World Health Organization Reporting System for Pancreaticobiliary Cytopathology: Review and Comparison to the Papanicolaou Society of Cytopathology System. Arch Pathol Lab Med 2025; 149:e39-e46. [PMID: 38190275 DOI: 10.5858/arpa.2023-0411-ra] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2023] [Indexed: 01/10/2024]
Abstract
CONTEXT.— The World Health Organization (WHO) Reporting System for Pancreaticobiliary Cytopathology (WHO System) is the product of a joint venture between the World Health Organization, the International Academy of Cytology, and the International Agency for Research on Cancer. The WHO System revises the Papanicolaou Society of Cytopathology System for Reporting Pancreaticobiliary Cytology (PSC System) and replaces the 6-tiered system with a 7-tiered system. OBJECTIVE.— To explain the WHO System and the differences with the PSC System. DATA SOURCES.— The WHO System and the PSC System of Reporting Pancreaticobiliary Cytopathology. CONCLUSIONS.— The diagnostic categories of the WHO System are "Insufficient/Inadequate/Nondiagnostic"; "Benign (Negative for Malignancy)"; "Atypical"; "Pancreaticobiliary Neoplasm, Low Risk/Low Grade (PaN-Low)"; "Pancreatic Neoplasm, High Risk/High Grade (PaN-High)"; "Suspicious for Malignancy"; and "Malignant." In the WHO System, the "benign" category includes both nonneoplastic and neoplastic lesions, so the "Neoplastic: Benign" category of the PSC system has been eliminated. Low-grade malignancies, pancreatic neuroendocrine tumors (PanNETs), and solid-pseudopapillary neoplasm (SPN) classified as "Neoplastic: Other" in the PSC System are classified as "Malignant" in the WHO System, leaving in the "Neoplasm" category intraductal lesions, which are divided into 2 new diagnostic categories: "Pancreaticobiliary Neoplasm (PaN)-Low Risk/Grade" and "PaN-High Risk/Grade." As with the PSC System, the WHO System advocates close correlation with imaging and encourages incorporation of ancillary testing into the final diagnosis, such as biochemical (carcinoembryonic antigen [CEA] and amylase) and molecular testing. The WHO System includes risk of malignancy per category, and reporting and diagnostic management options that recognize the variations in resources of low- and middle-income countries.
Collapse
Affiliation(s)
- Martha B Pitman
- From the Department of Pathology, Harvard Medical School, Boston, Massachusetts; and the Department of Pathology, Massachusetts General Hospital, Boston
| |
Collapse
|
32
|
Li P, Zhang H, Chen L, Gao X, Hu Y, Xu Q, Liu W, Chen W, Chen H, Yuan S, Wang M, Liu S, Dai M. Oral and fecal microbiota as accurate non-invasive tools for detection of pancreatic cancer in the Chinese population. Cancer Lett 2025; 612:217456. [PMID: 39800212 DOI: 10.1016/j.canlet.2025.217456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/02/2025] [Accepted: 01/10/2025] [Indexed: 01/15/2025]
Abstract
Pancreatic cancer (PCA), a leading cause of cancer-related deaths, has limited non-invasive diagnostic methods. We aimed to identify oral and fecal microbiome biomarkers and construct diagnostic classifiers. Oral and fecal samples from 97 PCA patients and 90 healthy controls underwent 16S rRNA sequencing. Samples were randomly divided into training and validation cohorts in a 7:3 ratio. Random forest models were constructed using training cohort and validated internally and externally in Chinese, Japanese, and Spanish populations. Results revealed significant dysbiosis of the oral and fecal microbiota of PCA patients. Most of the differential taxa shared between oral and fecal samples showed similar changes. Relative abundances of Streptococcus in oral samples, and of Bifidobacterium, Klebsiella and Akkermansia in fecal samples, were enriched in PCA. The fecal Firmicutes to Bacteroidota ratio was higher in PCA patient samples. Oral and fecal microbiome classifiers based on the top 20 contributing genera were constructed, and internal validation showed that the area under the curve (AUC) values were 0.963 and 0.890, respectively. The fecal microbiome classifier performed well in the external Chinese population, with an AUC of 0.878, but poorly in the Japanese and Spanish populations. Furthermore, fecal microbiomes could predict metastasis status in PCA patients, with an AUC of 0.804. In conclusion, oral and fecal microbiota were dysbiotic in PCA patients. Fecal microbiome classifier provides a feasible, non-invasive, and cost-effective tool with high precision for PCA screening in China; oral microbiome classifier requires further validation in external populations sampled with the same simple and convenient methods.
Collapse
Affiliation(s)
- Pengyu Li
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Hanyu Zhang
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Lixin Chen
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Xingyu Gao
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Ya Hu
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Qiang Xu
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Wenjing Liu
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Weijie Chen
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Haomin Chen
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Shuai Yuan
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Mingfei Wang
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Shili Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Cheelo College of Medicine, Shandong University, No.44 Wenhuaxi Road, Lixia District, Jinan, Shandong, 250012, China
| | - Menghua Dai
- Department of General Surgery, Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, No.1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
33
|
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
|
34
|
Mosalem OM, Abdelhakeem A, Abdel-Razeq NH, Babiker H. Pancreatic ductal adenocarcinoma (PDAC): clinical progress in the last five years. Expert Opin Investig Drugs 2025; 34:149-160. [PMID: 40012027 DOI: 10.1080/13543784.2025.2473698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 02/28/2025]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) remains a highly lethal malignancy with limited therapeutic options and poor overall survival. In recent years, advances in genomic profiling have revealed the complex molecular and cellular heterogeneity of PDAC, offering new avenues for therapeutic intervention. AREAS COVERED This review explores emerging therapeutic strategies targeting dysregulated molecular pathways, along with the tumor microenvironment, that have shown promise in overcoming drug resistance. Novel immunotherapy strategies, such as immune checkpoint inhibitors and CAR T-cell therapies, are currently being explored in an attempt to modulate PDAC immugnosuppressive microenvironment. Additionally, we highlight recent clinical trials over the last 5 years and innovative therapeutic strategies aiming to improve outcomes in PDAC. EXPERT OPINION Significant progress in genomic profiling, targeted therapies, and immunotherapy is shaping the treatment of PDAC. Despite challenges posed by its dense stroma and immune suppressive microenvironment, novel strategies such as IL 6 and CD137 inhibitors, CAR-T, and therapeutic cancer vaccines are promising. KRAS targeted therapies are expanding beyond G12C inhibitors, with novel drugs in development that will further improve treatment options. Additionally, tumor treating fields (TTF) are being investigated in locally advanced PDAC, with the PANOVA-3 trial potentially integrating this modality into future treatment strategies. Continued advancements in these areas will significantly enhance PDAC outcomes.
Collapse
Affiliation(s)
- Osama M Mosalem
- Department of Medicine, Division of Hematology Oncology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, USA
| | - Ahmed Abdelhakeem
- Department of Medicine, Division of Hematology Oncology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, USA
| | - Nayef H Abdel-Razeq
- Department of Medicine, Division of Hematology Oncology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, USA
| | | |
Collapse
|
35
|
Lasota M, Jankowski D, Wiśniewska A, Szeleszczuk Ł, Misterka-Kozaka A, Kaczor-Kamińska M, Zarzycka M, Patena M, Brzozowski T. Interaction of Avapritinib with Congo Red in Pancreatic Cancer Cells: Molecular Modeling and Biophysical Studies. Int J Mol Sci 2025; 26:1980. [PMID: 40076604 PMCID: PMC11901030 DOI: 10.3390/ijms26051980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Pancreatic cancer is a malignant tumor with one of the worst prognoses among solid tumors, characterized by resistance to treatment. Therefore, there is an urgent need for new methods of targeted therapy. Previous studies have shown that the overexpression of receptor tyrosine kinases such as c-KIT or PDGFR can increase proliferation, migration, and invasion of cancer cells. The aim of our study was to analyze aggregates between a supramolecular carrier (Congo red, CR) and a tyrosine kinase inhibitor (BLU-258) as well as to investigate the effect of the free inhibitor and its aggregate with Congo red (CR-BLU-258) on selected properties of pancreatic cells, including these cells' viability and three-dimensional cell spheroid cultures. To better understand the interactions between Congo red and BLU-258, we used molecular modeling in addition to biophysical methods. These attempts allowed us to determine the optimal molar ratio, which we used for in vitro studies on pancreatic cancer cell lines. A significantly greater decrease in the viability of the tested 3D cultures was observed after 48 h of incubation with CR-BLU-258, which resulted in a lower IC50 value for the tested co-aggregate compared with BLU-258 alone. Moreover, a higher resistance of PANC-1 and BxPC3 spheroid cells to the tested compounds was noted compared with the 2D culture model. A significantly lower response was observed in 3D cell cultures (BxPC3 and PANC-1) treated with BLU-258 alone compared with the 2D culture. Thus, our results showed that both BLU-258 (alone) and in its co-aggregate with Congo red exhibit anticancer activity, inhibiting the growth of pancreatic cancer cells and reducing their viability, survival, and migration. Both tested compounds also affected the phosphorylation of the selected signaling proteins. We conclude that the selected tyrosine kinase inhibitor (alone) and in its co-aggregate with Congo red exhibit anticancer activity and should be considered as a novel effective therapy against pancreatic cancer.
Collapse
Affiliation(s)
- Małgorzata Lasota
- Center for Biomedicine and Interdisciplinary Sciences, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Daniel Jankowski
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Anna Wiśniewska
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
| | - Łukasz Szeleszczuk
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097 Warsaw, Poland;
| | - Anna Misterka-Kozaka
- Center for Biomedicine and Interdisciplinary Sciences, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Marta Kaczor-Kamińska
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, 31-034 Krakow, Poland; (M.K.-K.); (M.Z.)
| | - Marta Zarzycka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, 31-034 Krakow, Poland; (M.K.-K.); (M.Z.)
| | - Maksym Patena
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Tomasz Brzozowski
- Center for Biomedicine and Interdisciplinary Sciences, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland
| |
Collapse
|
36
|
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
|
37
|
Marchese U, Pauly V, Pellat A, Richa Y, Fond G, Tzedakis S, Gaillard M, Fuchs B, Orleans V, Fuks D, El Amrani M, Boyer L. End-of-life care for patients with pancreatic cancer in France: a nationwide population-based cohort study. Ther Adv Med Oncol 2025; 17:17588359251320731. [PMID: 39990013 PMCID: PMC11843702 DOI: 10.1177/17588359251320731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
Background Pancreatic cancer, a frequently fatal disease with severe symptoms, can require high-intensity end-of-life (HI-EOL) care, posing challenges to patients' well-being. The examination of HI-EOL care to develop tailored interventions in the management of pancreatic cancer is a critical, yet underexplored area. Objectives The objective of this study was to assess the factors that influence the intensity of end-of-life (EOL) care in France. Design A retrospective study of patients registered in the French Nationwide database who were hospitalized in France for pancreatic adenocarcinoma from January 1, 2014 to December 31, 2019, and subsequently died during the follow-up period. Methods Data on patient demographics, clinical characteristics, hospitalization details, and palliative care were collected. The primary outcome measure was the evaluation of HI-EOL care, defined by indicators such as death in an intensive care unit (ICU), multiple hospitalizations, and chemotherapy administration within the last 30 days of life. Secondary outcomes included indicators of most-intensive EOL (MI-EOL) care and invasive procedures (IP). Univariate and multivariate logistic regression analyses were conducted to identify factors associated with each outcome measure. Results A total of 42,696 patients who died from pancreatic adenocarcinoma were included. Among them, 41.1% experienced HI-EOL, with the most common indicators being multiple hospitalizations and death in an ICU, emergency room, or acute care unit. A smaller proportion (2.8%) received MI-EOL care, while 28.1% underwent IPs in the last 30 days of life. The multivariate analysis revealed that male gender and follow-up in non-cancer specialized care facilities were associated with a higher risk of HI-EOL. Conversely, palliative care involvement and older age at death were identified as protective factors. Male gender, older age at death, and palliative care involvement were associated with lower rates of MI-EOL care and IPs. Conclusion These results underscore the importance of palliative care integration and individualized approaches in improving the EOL quality of care and patient outcomes for individuals with advanced pancreatic cancer.
Collapse
Affiliation(s)
- Ugo Marchese
- Department of Digestive, HPB and Endocrine Surgery, Hôpital Cochin, AP-HP Centre, 27 rue du Faubourg St Jacques, Paris 75014, France
- Université de Paris Cité, 15 rue de l’école de médecine 75006 Paris, France
| | - Vanessa Pauly
- Département d’Information Médicale, Hôpital de la Timone, 264 Rue Saint-Pierre, 13005 Marseille, France
- Université d’Aix-Marseille, Jardin du Pharo, 58 Boulevard Charles Livon, 13007 Marseille, France
| | - Anna Pellat
- Department of Gastroenterology and Digestive Oncology, Hôpital Cochin, AP-HP Centre, 27 rue du Faubourg St Jacques, 75014 Paris, France
- Université de Paris Cité, 15 rue de l’école de médecine 75006 Paris, France
| | - Yasmina Richa
- School of Medicine, University College Cork, Cork, Ireland
| | - Guillaume Fond
- Department of University Psychiatry, Sainte Marguerite University Hospital, Assistance Publique des Hôpitaux de Marseille, Marseille, France
- Assistance Publique des Hôpitaux de Marseille, Aix-Marseille University, UR3279: Health Service Research and Quality of Life Center - CEReSS, Marseille, France
| | - Stylianos Tzedakis
- Department of Digestive, HPB and Endocrine Surgery, Hôpital Cochin, AP-HP Centre, 27 rue du Faubourg St Jacques, 75014 Paris, France
- Université de Paris Cité, 15 rue de l’école de médecine 75006 Paris, France
| | - Martin Gaillard
- Department of Digestive, HPB and Endocrine Surgery, Hôpital Cochin, AP-HP Centre, 27 rue du Faubourg St Jacques, 75014 Paris, France
- Université de Paris Cité, 15 rue de l’école de médecine 75006 Paris, France
| | - Basile Fuchs
- Département d’Information Médicale, Hôpital de Brest, 2 avenue Foch, 29200 Brest, France
| | - Veronica Orleans
- Département d’Information Médicale, Hôpital de la Timone, 264 Rue Saint-Pierre, 13005 Marseille, France
- Université d’Aix-Marseille, Jardin du Pharo, 58 Boulevard Charles Livon, 13007 Marseille, France
| | - David Fuks
- Department of Digestive Surgery, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland
| | - Mehdi El Amrani
- Department of Digestive Surgery and Transplantation, CHRU de Lille, 2 Av. Oscar Lambret, 59000 Lille, France
- Université de Lille, 42 Rue Paul Duez, 59000 Lille, France
| | - Laurent Boyer
- Département d’Information Médicale, Hôpital de la Timone, 264 Rue Saint-Pierre, 13005 Marseille, France
- Université d’Aix-Marseille, Jardin du Pharo, 58 Boulevard Charles Livon, 13007 Marseille, France
| |
Collapse
|
38
|
Frimpong E, Annor E, Bulusu R, Okoro J, Kiros GE, Reams R, Agyare E. Sociodemographic characteristics associated with pancreatic cancer incidence and mortality among Blacks in the United States: a SEER-based study. Am J Cancer Res 2025; 15:705-722. [PMID: 40084357 PMCID: PMC11897636 DOI: 10.62347/gjcx1238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/03/2025] [Indexed: 03/16/2025] Open
Abstract
Pancreatic cancer (PC) is the third leading cause of all cancer-related fatalities and accounts for approximately 3% of cancer cases in the United States. PC survival rates are lower in Blacks compared to other races, and this has been attributed to socioeconomic and genetic factors. In this study, we evaluated sociodemographic and genetic characteristics associated with PC incidence and mortality among Blacks. Data from the SEER 22 registries (2000-2020) were used to calculate the incidence rates and relative survival. County mortality rates from 2017 to 2021 were analyzed. Incidence rate ratios based on gender, age, primary disease site, stage, level of education, and poverty were calculated. Survival analysis was conducted using the Kaplan-Meier method. Mutant gene expression was obtained from the MSK-CHORD tumor registry. Overall, 48,606 Black patients were diagnosed with malignant PC between 2000 and 2020: females (53.53%) and males (46.47%). Both males and females experienced a slight increase in Annual Percent Change (APC) of PC incidence (0.24, 95% CI, -0.02-0.53) and (0.22, 95% CI, -0.05-0.51), respectively, from 2000 to 2020. Males aged 55 to 75 years were most frequently affected. Overall incidence risk from 2000-2020 by age was higher in Black males IRR > 1 (1.18, 95% CI, 1.16-1.21). The most common primary PC site for Black males and females was the head of the pancreas, 49.06% and 49.88%, respectively. By staging, distant PC had the highest frequency in Blacks. Poverty level was associated with PC incidence among females and PC mortality among both males and females. Stage was associated with survival among males with localized and regional PC. The 5-year relative survival was less than 11% across combined PC stages for both sexes. Black males had a relatively lower 5-year survival than Black females in localized (31.7 vs. 37.2%) and distant PC (2.6% vs. 2.90%). Mutant KRAS expression was higher in Black males. PC incidence and mortality were significantly higher in Black males. Our analysis points to the importance of poverty alleviation programs that target females are likely to reduce PC incidence. Furthermore, receiving recommended screening for PC and early-stage diagnostics is important to lower PC mortality.
Collapse
Affiliation(s)
- Esther Frimpong
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, The United States
| | - Eugene Annor
- Department of Internal Medicine, University of Illinois College of Medicine at PeoriaPeoria, Illinois, The United States
| | - Raviteja Bulusu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, The United States
| | - Joy Okoro
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, The United States
| | - Gebre-Egziabher Kiros
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, The United States
| | - Renee Reams
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, The United States
| | - Edward Agyare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, The United States
| |
Collapse
|
39
|
Qing Q, Deng X, Deng X, Mou T, Li B, Tan Y, Wu Q. A single-center study examining the safety and effectiveness of ERCP with pancreatoscopy and endoluminal radiofrequency ablation for main-duct IPMN treatment. Sci Rep 2025; 15:5420. [PMID: 39948142 PMCID: PMC11825723 DOI: 10.1038/s41598-025-89889-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: 07/09/2024] [Accepted: 02/10/2025] [Indexed: 02/16/2025] Open
Abstract
To assess the safety and efficacy of treatment for intraductal papillary mucinous neoplasm (IPMN) involving the main pancreatic duct (MPD) using endoscopic retrograde cholangiopancreatography (ERCP) combined with digital single-operator pancreatoscopy and radiofrequency ablation (RFA). A retrospective analysis was performed. Patients with episodes of acute pancreatitis at the First Affiliated Hospital of Chongqing Medical University were screened. When the diagnosis of IPMN was confirmed by ERCP with pancreatoscopy, patients who underwent endoluminal RFA treatment were enrolled. The most frequent complications were monitored to assess the safety of treatment. Disease progression and recurrence of pancreatitis symptoms were regarded as the indicators of treatment efficacy. In total, 50 patients were screened, and 14 patients who undertook the lesion ablation using RFA were enrolled. Mild adverse events occurred, while they were relieved under symptomatic treatment. No severe complications were recorded. All patients received regular follow-ups, during which an episode of IPMN-related pancreatitis occurred in 1 patient, and disease progression was found in 1 patient who successively underwent pancreaticoduodenectomy. Regional treatment of IPMN lesions in the MPD exhibited satisfactory effect in preventing lesion progression and relieving pancreatitis symptoms through the combined use of ERCP with pancreatoscopy and RFA, without causing severe complications.
Collapse
Affiliation(s)
- Qiang Qing
- Department of Hepatobiliary Surgery, Ziyang People's Hospital, Ziyang, 641300, Sichuan, China
| | - Xiang Deng
- Department of General Surgery, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China
| | - Xin Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Tong Mou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Bin Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ye Tan
- Chongqing University Fuling Hospital, Chongqing, 408000, China.
- Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, 610041, Sichuan, China.
| | - Qiao Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
40
|
Baker JG, Sloan EK, Pfleger KDG, McCormick PJ, Salmerón C, Insel PA. Pancreatic Ductal Adenocarcinoma, β-blockers, and Antihistamines: A Clinical Trial Is Needed. FUNCTION 2025; 6:zqae050. [PMID: 39547938 PMCID: PMC11815576 DOI: 10.1093/function/zqae050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024] Open
Affiliation(s)
- Jillian G Baker
- Cell Signalling, School of Life Sciences, C Floor Medical School, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
- Respiratory Medicine, Queen’s Medical Centre, Nottingham University Hospitals, Nottingham NG7 2UH, UK
| | - Erica K Sloan
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Science, Monash University, Parkville Victoria 3052, Australia
| | - Kevin D G Pfleger
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Peter J McCormick
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 3GE, UK
| | - Cristina Salmerón
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Paul A Insel
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
41
|
Song Y, Gao S, Jiang J, Zhang Y, Zhang J, Wang X, Lv L, Zhou Z, Wang J. Inhibition Effects and Mechanism Study of rAj-HRP30, a Recombinant Histidine-Rich Peptide from Apostichopus japonicus, on the Viability of Pancreatic Ductal Adenocarcinoma Cells Panc01 and Panc02. Int J Mol Sci 2025; 26:1485. [PMID: 40003950 PMCID: PMC11854995 DOI: 10.3390/ijms26041485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/05/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
rAj-HRP30 is a recombinant peptide derived from the wild-type rAj-HRP of Apostichopus japonicus through a gene-shortening mutation. It has a high histidine content (53.3% in its primary structure) and a molecular weight of 3.919 kDa, classifying it as a histidine-rich peptide. The literature reports indicate that human histidine-rich peptides exhibit antitumor activity. Previous research by our group demonstrated similar properties in rAj-HRP, the precursor of rAj-HRP30. Therefore, this study used Panc01 (human) and Panc02 (mouse) cells-highly malignant models with limited targeted therapies-to investigate the antitumor activity and mechanisms of rAj-HRP30 and evaluate its potential for pancreatic cancer treatment. This study designed a gene-shortening strategy for rAj-HRP and artificially synthesized the gene sequence of rAj-HRP30. The cDNA sequence of rAj-HRP30 was cloned into the pET23b vector, and the recombinant plasmid pET23b-HRP30 was transformed into E. coli BL21 for expression. Following IPTG induction, the recombinant peptide was purified using nickel ion affinity chromatography, yielding rAj-HRP30 with a purity exceeding 95%. rAj-HRP30 markedly inhibited the adhesion, migration, and invasion of Panc01 and Panc02 cells. It also disrupted cellular morphology and cytoskeletal structure while inducing apoptosis. These effects were dose-dependent. After confirming the in vitro anticancer activity of rAj-HRP30, this study employed Panc02 cells as a model to investigate its inhibitory mechanisms using Western blot analysis. The results revealed that rAj-HRP30 reduced FGFR1 expression in Panc02 cells and inhibited the downstream FYN and FAK signaling pathways, subsequently blocking the PI3K/AKT signaling and apoptosis pathways. In the apoptotic pathway, rAj-HRP30 was able to downregulate the expression of Bcl-2, Caspase-9, Caspase-3, Caspase-7, and PARP1 and upregulate the expression of Bax, cleaved Caspase-9, cleaved Caspase-3, cleaved Caspase-7, and cleaved-PARP1 to induce apoptosis in Panc02 cells. Furthermore, rAj-HRP30 also downregulated the expression of MMP2 and MMP9, thereby inhibiting the migration and invasion of Panc02 cells. Conclusion: rAj-HRP30 exhibits significant inhibitory effects on pancreatic ductal adenocarcinoma Panc01 and Panc02 cells in vitro. Its mechanism involves FGFR1-related signaling and apoptosis pathways. rAj-HRP30 shows promise as a therapeutic agent targeting FGFR for pancreatic cancer.
Collapse
Affiliation(s)
- Yuyao Song
- Ministry of Agriculture and Rural Affairs Key Lab of Protection and Utilization of Aquatic Germplasm Resource, Liaoning Province Key Lab of Germplasm Improvement and Fine Seed Breeding of Marine Aquatic Animals, Liaoning Ocean and Fisheries Science Research Institute, Dalian 116023, China; (Y.S.); (S.G.); (J.J.)
- School of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Y.Z.); (J.Z.); (X.W.)
| | - Shan Gao
- Ministry of Agriculture and Rural Affairs Key Lab of Protection and Utilization of Aquatic Germplasm Resource, Liaoning Province Key Lab of Germplasm Improvement and Fine Seed Breeding of Marine Aquatic Animals, Liaoning Ocean and Fisheries Science Research Institute, Dalian 116023, China; (Y.S.); (S.G.); (J.J.)
| | - Jingwei Jiang
- Ministry of Agriculture and Rural Affairs Key Lab of Protection and Utilization of Aquatic Germplasm Resource, Liaoning Province Key Lab of Germplasm Improvement and Fine Seed Breeding of Marine Aquatic Animals, Liaoning Ocean and Fisheries Science Research Institute, Dalian 116023, China; (Y.S.); (S.G.); (J.J.)
| | - Yuebin Zhang
- School of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Y.Z.); (J.Z.); (X.W.)
| | - Jingyu Zhang
- School of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Y.Z.); (J.Z.); (X.W.)
| | - Xiaona Wang
- School of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Y.Z.); (J.Z.); (X.W.)
| | - Li Lv
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China;
| | - Zunchun Zhou
- Ministry of Agriculture and Rural Affairs Key Lab of Protection and Utilization of Aquatic Germplasm Resource, Liaoning Province Key Lab of Germplasm Improvement and Fine Seed Breeding of Marine Aquatic Animals, Liaoning Ocean and Fisheries Science Research Institute, Dalian 116023, China; (Y.S.); (S.G.); (J.J.)
| | - Jihong Wang
- School of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Y.Z.); (J.Z.); (X.W.)
| |
Collapse
|
42
|
Zhou Y, Tang Y, Huang F, Wang Z, Wen Z, Fang Q, Wang C. The miR-1305/KLF5 negative regulatory loop affects pancreatic cancer cell proliferation and apoptosis. Hum Cell 2025; 38:51. [PMID: 39921786 DOI: 10.1007/s13577-025-01173-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 01/05/2025] [Indexed: 02/10/2025]
Abstract
Pancreatic cancer (PC) is characterized by a high relapse rate and unfavorable prognosis. Currently, the optimal treatment for PC is complete resection followed by adjuvant systemic chemotherapy. Nevertheless, tumor cell repopulation and subsequent tumor relapse and metastasis after chemotherapy result in a poor prognosis. Therefore, it is of great value to explore the potential molecular mechanisms underlying PC for developing novel treatment strategies. Herein, we aimed to investigate the potential regulatory mechanism of miR-1305 upon aerobic proliferation, metastasis, and apoptosis in PC. miR-1305 was downregulated in PC tissues and cell lines. miR-1305 overexpression prominently inhibited PC cell proliferation and metastasis promoted cell apoptosis in vitro, and alleviated PC formation in vivo. As predicted, KLF5 could directly bind to miR-1305. Silencing of KLF5 or KLF5 inhibitor (ML264) suppressed PC cell viability and cell invasion, and enhanced cell apoptosis. KLF5 restrained miR-1305 transcription and expression by binding to its promoter region. miR-1305 exerted a suppressive effect on PC cell proliferation and apoptosis via regulation of the KLF5-ERBB2 axis; KLF5 gene is a transcriptional regulator of miR-1305, promising to be a new target for the diagnosis and treatment of PC.
Collapse
Affiliation(s)
- Yufu Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, China
| | - Yulin Tang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, China
| | - Feizhou Huang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, China
| | - Zhichao Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, China
| | - Zhengbin Wen
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, China
| | - Qi Fang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, China
| | - Changfa Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan Province, China.
| |
Collapse
|
43
|
Chen G, Li B, Li T, Lin M, Zhong H, Xie X, Zhang Q, Chen Q, Meng X, Xiao Z, Shuai X. Core-Satellite Nanoassembly Overcomes Spatial Heterogeneity of Dendric Cell Distribution in Pancreatic Tumors for Effective Chemoimmunotherapy. ACS NANO 2025; 19:4739-4753. [PMID: 39834130 DOI: 10.1021/acsnano.4c15444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Pancreatic cancer therapies such as chemotherapy and immunotherapy are hindered by the dense extracellular matrix known as physical barriers, leading to heterogeneity impeding the effective penetration of chemotherapeutic agents and activation of antitumor immune responses. To address this challenge, we developed a hybrid nanoassembly with a distinct core-satellite-like heterostructure, PLAF@P/T-PD, which is responsive to both internal pH/redox and external ultrasound stimulations. This heterostructural nanoassembly features a polymersome core encapsulating an ultrasound contrast agent perfluoropentane and a chemotherapeutic agent Taxol (PLAF@P/T) electrostatically coated with satellite-like polyplexes carrying an immune agonist dsDNA (PD), which brings about synergistic functions inside the pancreatic tumor. The PLAF@P/T core functions as an enhancer for intratumor delivery through size enlargement and charge conversion in response to reactive oxygen species (ROS) and low pH, which triggers polyplex release and enables ultrasound-assisted tumor-penetrating Taxol delivery. Meanwhile, the released cationic polyplexes function as nucleic nanomedicine preferentially engulfed by peripheral dendritic cells (DCs) for immune modulation. Animal studies in mouse orthotopic pancreatic tumor model demonstrated exceptional therapeutic efficacy against both primary and metastatic tumors, which underlines the potential of this heterostructural nanoplatform for overcoming the therapeutic challenges associated with the heterogeneous physical barrier hindering intratumor drug delivery in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Gengjia Chen
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- Department of Radiology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Bo Li
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Tan Li
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Minzhao Lin
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Huihai Zhong
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Xiaoxue Xie
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Qiaoyun Zhang
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Qi Chen
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaochun Meng
- Department of Radiology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
44
|
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
|
45
|
Balaraman AK, Moglad E, Afzal M, Babu MA, Goyal K, Roopashree R, Kaur I, Kumar S, Kumar MR, Chauhan AS, Hemalatha S, Gupta G, Ali H. Liquid biopsies and exosomal ncRNA: Transforming pancreatic cancer diagnostics and therapeutics. Clin Chim Acta 2025; 567:120105. [PMID: 39706249 DOI: 10.1016/j.cca.2024.120105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Pancreatic cancer is a highly fatal malignancy due to poor early detection rate and resistance to conventional therapies. This review examines the potential for liquid biopsy as a transformative technology to identify diagnostic and therapeutic targets in pancreatic cancer. Specifically, we explore emerging biomarkers such as exosomal non-coding RNAs (ncRNAs), circulating tumor DNA (ctDNA), and circulating tumor cells (CTCs). Tumor-derived exosomes contain nucleic acid and protein that reflect the unique molecular landscape of the malignancy and can serve as an alternative diagnostic approach vs traditional biomarkers like CA19-9. Herein we highlight exosomal miRNAs, lncRNAs, and other ncRNAs alongside ctDNA and CTC-based strategies, evaluating their combined ability to improve early detection, disease monitoring and treatment response. Furthermore, the therapeutic implications of ncRNAs such as lncRNA UCA1 and miR-3960 in chemoresistance and progression are also discussed via suppression of EZH2 and PTEN/AKT pathways. Emerging therapeutic strategies that target the immune response, epithelial-mesenchymal transition (EMT) and drug resistance are explored. This review demonstrates a paradigm shift in pancreatic cancer management toward personalized, less invasive and more effective approaches.
Collapse
Affiliation(s)
- Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - MRavi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research and Innovation, Uttaranchal University, India
| | - S Hemalatha
- Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Porur, Chennai, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| |
Collapse
|
46
|
Bonazzi VF, Aoude LG, Brosda S, Bradford JJ, Lonie JM, Loffler KA, Gartside MG, Patel K, Mukhopadhyay P, Keane C, Gebski V, Kench JG, Goldstein D, Waddell N, Barbour AP. C-reactive protein is a prognostic biomarker in pancreatic ductal adenocarcinoma patients. Asia Pac J Clin Oncol 2025; 21:77-86. [PMID: 37415393 PMCID: PMC11733851 DOI: 10.1111/ajco.13993] [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: 12/01/2021] [Revised: 04/04/2023] [Accepted: 06/26/2023] [Indexed: 07/08/2023]
Abstract
AIM The 5-year survival rate of pancreatic ductal adenocarcinoma (PDAC) is approximately 11% and has only improved marginally over the last three decades. For operable PDAC, resection and adjuvant FOLFIRINOX chemotherapy is standard of care. There is growing interest in perioperative regimens to improve outcomes. The non-randomized Phase II study "Gemcitabine and Abraxane for resectable Pancreatic cancer" (GAP) demonstrated the feasibility of perioperative gemcitabine/abraxane. Long-term survival in PDAC requires an effective immune response; hence, we undertook this translational study of the GAP trial cohort to identify immune-oncology biomarkers for clinical use. METHODS We combined Nanostring nCounter technology with immunohistochemistry to investigate the correlation between gene expression and overall patient survival. Findings were investigated in samples from the International Cancer Genome Consortium (ICGC, n = 88) and the Australian Pancreatic Genome Initiative (APGI, n = 227). RESULTS We confirmed that human equilibrative nucleoside transporter 1 (hENT1) expression was not a prognostic marker in PDAC but patients with high levels of hENT1 were more likely to live longer than 24 months post-surgery. Additionally, CD274 (PD-L1) and two novel biomarkers of survival, cathepsin W (CTSW) and C-reactive protein (CRP), were identified in the GAP cohort (n = 19). CRP expression was confirmed in data from the ICGC. Although PD-L1 and CTSW proteins were not significant across all three cohorts, results show that low CRP mRNA and protein expression are associated with longer overall survival in all three patient groups. CONCLUSION PDAC patients with long survival have higher hENT1 expression levels. Furthermore, CRP expression is a biomarker of poor prognosis following perioperative chemotherapy and resection in PDAC patients and thus may be useful for identifying patients who could benefit from more aggressive adjuvant strategies.
Collapse
Affiliation(s)
- Vanessa F. Bonazzi
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Lauren G. Aoude
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Sandra Brosda
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Julia J. Bradford
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - James M. Lonie
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Kelly A. Loffler
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
- College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Michael G. Gartside
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Kalpana Patel
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | | | - Colm Keane
- Mater Research Institute‐UQSouth BrisbaneQueenslandAustralia
| | - Val Gebski
- NHMRC Clinical Trials CentreCamperdownNew South WalesAustralia
| | - James G. Kench
- Royal Prince Alfred HospitalCamperdownNew South WalesAustralia
- University of Sydney Central Clinical SchoolCamperdownNew South WalesAustralia
| | - David Goldstein
- University of NSW Prince of Wales Clinical SchoolRandwickNew South WalesAustralia
| | - Nicola Waddell
- QIMRBerghofer Medical Research InstituteHerstonQueenslandAustralia
| | - Andrew P. Barbour
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
- Princess Alexandra HospitalWoolloongabbaQueenslandAustralia
| | | |
Collapse
|
47
|
Liang YY, Lu Z, Liu HW, Huang Q, Zheng XT, Li XA, Zhou Y. Anti-tumor effects of nanosecond pulsed electric fields in a murine model of pancreatic cancer. Bioelectrochemistry 2025; 161:108803. [PMID: 39241512 DOI: 10.1016/j.bioelechem.2024.108803] [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: 01/03/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024]
Abstract
Nanosecond Pulsed Electric Fields (nsPEFs) treatment has demonstrated anti-tumor effects on various cancer cell lines. However, the use of this treatment in pancreatic cancer is limited. This study demonstrated that nsPEFs treatment effectively suppressed the proliferation and metastasis of pancreatic cancer cells, while also inducing DNA damage. Meanwhile, animal experiments have shown that nsPEFs effectively suppressed the growth of pancreatic cancer, even in cases where the tumor volume exceeded 500-600 mm3 at the initiation of treatment. Notably, a single treatment session was found to significantly inhibit tumor growth, while also showing no adverse effects on the main organs of the mice. RNA sequencing and bioinformatics revealed that seven key genes (CDK1, CENPA, UBE2C, CCNB2, PLK1, CCNA2, and CCNB14) were significantly correlated with the overall survival rate of patients with pancreatic cancer. Through the application of the competing endogenous RNA (ceRNA) hypothesis, two miRNAs (has-let-7b-5p and hsa-miR-193b-3p) and four lncRNAs (MIR4435-2HG, ZNF436-AS1, LINC01089, and MIR4435-2HG) were identified as significantly impacting the overall survival of pancreatic cancer patients. We have effectively developed an mRNA-miRNA-lncRNA network that has the potential to stimulate further investigation into the underlying mechanisms of nsPEFs on pancreatic cancer.
Collapse
Affiliation(s)
- Yuan-Yuan Liang
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang 621000, Sichuan, China
| | - Zhou Lu
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang 621000, Sichuan, China
| | - Hong-Wei Liu
- Institute of Fluid Physics, CAEP, Mianyang 621000, China
| | - Qi Huang
- Xinjiang University of Science & Technology, Xinjiang 830000, China
| | - Xue-Ting Zheng
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang 621000, Sichuan, China
| | - Xiao-An Li
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang 621000, Sichuan, China.
| | - Yan Zhou
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang 621000, Sichuan, China.
| |
Collapse
|
48
|
Sheng T, Dokmanovich B, Yu Y, Liu C, Fan H, Prizment AE, Anderson KE, Zhang J. Associations Between Intakes of Carotenoids and Tocopherols with Risk of Pancreatic Cancer in a Population-Based Case-Control Study. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2025; 44:128-136. [PMID: 39302853 PMCID: PMC11757089 DOI: 10.1080/27697061.2024.2404578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVE Experimental studies suggest that carotenoids and tocopherols modulate pancreatic carcinogenesis because they have antioxidant and other functions. We investigated the associations between intakes of these compounds and the risk of pancreatic cancer in a case-control study conducted in 1994-1998. METHODS The present analysis included 150 cases of pancreatic cancer recruited from all hospitals in the metropolitan area of the Twin Cities and Mayo Clinic and 459 controls randomly selected from the general population and frequency matched to cases by age, sex, and race. The intakes of carotenoids and tocopherols were assessed with a validated food frequency questionnaire. Unconditional logistic regression analysis was performed to evaluate the associations of interest. RESULTS The energy-adjusted intake of lutein/zeaxanthin was significantly lower in cases (2410 µg/day) than in controls (3020 µg/day). After adjustment for confounders, persons in the fourth quartile of lutein/zeaxanthin intake had a reduced risk of pancreatic cancer compared with those in the first quartile [odds ratio (OR) (95% CI): 0.40 (0.17-0.91)]. There were no significant associations with intakes of other carotenoids and tocopherols considered and with a composite score created from all individual carotenoids examined. We did not detect any significant interactions of intakes of carotenoids and tocopherols with age, sex, cigarette smoking, or alcohol intake in relation to pancreatic cancer risk. CONCLUSION The present study suggests an inverse association between lutein/zeaxanthin intake and pancreatic cancer risk, but a potential beneficial effect was not observed for other carotenoids and tocopherols.
Collapse
Affiliation(s)
- Tianchen Sheng
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
| | - Bronson Dokmanovich
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
| | - Yunpeng Yu
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
- Department of Biostatistics and Health Data Science, Indiana University Richard M. Fairbanks School of Public Health and School of Medicine, Indianapolis, IN, USA
| | - Chunliang Liu
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
- Department of Gastroenterology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Hao Fan
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health, Bloomington, IN, USA
| | - Anna E. Prizment
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Kristin E. Anderson
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Jianjun Zhang
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| |
Collapse
|
49
|
Beljkas M, Ruzic D, Djuric A, Vuletic A, Tchiehe GN, Jallet C, Cadet-Daniel V, Arimondo PB, Santibanez JF, Srdic-Rajic T, Nikolic K, Oljacic S, Petkovic M. Pioneering first-in-class HDAC-ROCK inhibitors as potential multitarget anticancer agents. Future Med Chem 2025; 17:393-407. [PMID: 39885716 PMCID: PMC11834526 DOI: 10.1080/17568919.2025.2459589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/22/2025] [Indexed: 02/01/2025] Open
Abstract
AIM With the aim of simultaneously modulating the epigenetic system and the protein kinase pathway, we selected the enzyme histone deacetylase (HDAC) and the Rho-associated protein kinases (ROCK) as desired targets to develop potential multitarget anticancer agents with additional antimetastatic properties. We report here the rational design, synthesis, and biological evaluation of the first-in-class HDAC/ROCK multitarget inhibitors in pancreatic ductal adenocarcinoma (PDAC) and triple-negative breast cancer (TNBC). MATERIALS AND METHODS A molecular docking study performed with the Gold software was used to develop HDAC/ROCK multitarget inhibitors. IC50 values were determined by enzyme assays. The cytotoxicity, anti-migratory and anti-invasive properties of the inhibitors were evaluated using triple-negative breast cancer cells (MDA-MB-231 and HCC 1973) and pancreatic ductal adenocarcinoma cells (Panc-1 and MiaPaCa-2). RESULTS C-9 showed significant inhibition of HDAC6, ROCK1 and ROCK2. At the same time, this compound showed strong antiproliferative effects on MDA-MB-231, MiaPaCa-2 and Panc-1 cell lines with IC50 values of 5.81 μM, 3.87 μM and 19.57 μM. In addition, it demonstrated great anti-invasive and anti-migratory effects. CONCLUSION The findings of this study strongly suggest that the simultaneous inhibition of ROCK and HDACs holds significant potential as a promising therapeutic strategy in the advancement of cancer treatment.
Collapse
Affiliation(s)
- Milan Beljkas
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Dusan Ruzic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Ana Djuric
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Ana Vuletic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Guilaine Nchugoua Tchiehe
- Department of Structural Biology and Chemistry, Epigenetic Chemical Biology, Institut Pasteur, Université Paris Cité, CNRS UMR3523 Chem4Life, Department of Structural Biology and Chemistry, Paris, France
| | - Corinne Jallet
- Department of Structural Biology and Chemistry, Epigenetic Chemical Biology, Institut Pasteur, Université Paris Cité, CNRS UMR3523 Chem4Life, Department of Structural Biology and Chemistry, Paris, France
| | - Véronique Cadet-Daniel
- Department of Structural Biology and Chemistry, Epigenetic Chemical Biology, Institut Pasteur, Université Paris Cité, CNRS UMR3523 Chem4Life, Department of Structural Biology and Chemistry, Paris, France
| | - Paola B. Arimondo
- Department of Structural Biology and Chemistry, Epigenetic Chemical Biology, Institut Pasteur, Université Paris Cité, CNRS UMR3523 Chem4Life, Department of Structural Biology and Chemistry, Paris, France
| | - Juan F. Santibanez
- Group for Molecular Oncology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tatjana Srdic-Rajic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Slavica Oljacic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Milos Petkovic
- Department of Organic Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
50
|
Wik SL, Tian W, Zhong CC, Sawhney A, Gao M, Yu Q, Xue F, Chan SC, Chow SH, Adebisi YA, Yuan J, Lucero‐Prisno DE, Wong MCS, Huang J. Distribution, Risk Factors and Epidemiological Trends of Pancreatic Cancer Across Countries' Income Levels: A Comprehensive Analysis. Cancer Rep (Hoboken) 2025; 8:e70154. [PMID: 39957387 PMCID: PMC11830997 DOI: 10.1002/cnr2.70154] [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] [Revised: 01/03/2025] [Accepted: 01/31/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND Globally, pancreatic cancer poses a significant concern for public health. AIMS The objective of this study was to assess the burden of pancreatic cancer on varying income levels. METHODS AND RESULTS Data from the Global Burden of Disease Study (GBD) 2021 and Gross Domestic Product Per Capita data were utilised in this study. All countries were categorised into four groups based on their income levels. Age-standardised incidence, mortality and disability-adjusted life years (DALYs) rates were the primary parameters to analyse the burden of pancreatic cancer. The associations between pancreatic cancer burden and countries' economic levels were analysed with linear regression models. High-income-level countries generally had a higher burden compared to other income levels in 2021. Greenland had the highest rate of age-standardised DALYs at 374.93 per 100 000, followed by Uruguay (297.06) and Monaco (290.87). A higher gross domestic product (GDP) per capita was linked to a higher age-standardised incidence (β = 0.77, 95% CI = 0.63, 0.90, p < 0.001), mortality (β = 0.72, 95% CI = 0.59, 0.86, p < 0.001) and DALYs (β = 14.59, 95% CI = 11.38, 17.80, p < 0.001). From 1990 to 2021, the pancreatic cancer burden increased across all income levels, with the most pronounced rise seen in lower-middle-income countries. Smoking-related age-standardised DALYs have decreased since 1990. However, there was a notable increase in males in upper-middle-income countries during the same period. CONCLUSION In conclusion, the pancreatic cancer burden has been increasing globally. The burden of pancreatic cancer varies significantly among countries with different income levels. Effective preventions are needed to control the burden of pancreatic cancer.
Collapse
Affiliation(s)
- Sofia Laila Wik
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, the Chinese University of Hong KongHong Kong SARChina
- Karolinska InstituteSolnaSweden
| | - Wenxin Tian
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, the Chinese University of Hong KongHong Kong SARChina
| | - Claire Chenwen Zhong
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, the Chinese University of Hong KongHong Kong SARChina
- Centre for Health Education and Health Promotion, Faculty of Medicine, the Chinese University of Hong KongHong Kong SARChina
| | - Apurva Sawhney
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, the Chinese University of Hong KongHong Kong SARChina
| | - Mingjun Gao
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, the Chinese University of Hong KongHong Kong SARChina
- Adam Smith Business School, College of Social Science, University of GlasgowGlasgowUK
| | - Qinyao Yu
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, the Chinese University of Hong KongHong Kong SARChina
- Jinan University‐University of Birmingham Joint Institute, Jinan UniversityGuangzhouChina
- School of Mathematics, College of Engineering and Physical Sciences, University of BirminghamBirminghamUK
| | - Fanyu Xue
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, the Chinese University of Hong KongHong Kong SARChina
- Faculty of Health Sciences, University of OttawaOttawaCanada
| | - Sze Chai Chan
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, the Chinese University of Hong KongHong Kong SARChina
| | - Shui Hang Chow
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, the Chinese University of Hong KongHong Kong SARChina
| | | | - Jinqiu Yuan
- Clinical Research Center, Big Data Center, the Seventh Affiliated Hospital, Sun Yat‐Sen UniversityShenzhenGuangdongChina
| | - Don Eliseo Lucero‐Prisno
- Department of Global Health and DevelopmentLondon School of Hygiene and Tropical MedicineLondonUK
| | - Martin C. S. Wong
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, the Chinese University of Hong KongHong Kong SARChina
- Centre for Health Education and Health Promotion, Faculty of Medicine, the Chinese University of Hong KongHong Kong SARChina
| | - Junjie Huang
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, the Chinese University of Hong KongHong Kong SARChina
- Centre for Health Education and Health Promotion, Faculty of Medicine, the Chinese University of Hong KongHong Kong SARChina
| | | |
Collapse
|