1
|
Murray K, Oldfield L, Stefanova I, Gentiluomo M, Aretini P, O'Sullivan R, Greenhalf W, Paiella S, Aoki MN, Pastore A, Birch-Ford J, Rao BH, Uysal-Onganer P, Walsh CM, Hanna GB, Narang J, Sharma P, Campa D, Rizzato C, Turtoi A, Sever EA, Felici A, Sucularli C, Peduzzi G, Öz E, Sezerman OU, Van der Meer R, Thompson N, Costello E. Biomarkers, omics and artificial intelligence for early detection of pancreatic cancer. Semin Cancer Biol 2025; 111:76-88. [PMID: 39986585 DOI: 10.1016/j.semcancer.2025.02.009] [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: 11/29/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is frequently diagnosed in its late stages when treatment options are limited. Unlike other common cancers, there are no population-wide screening programmes for PDAC. Thus, early disease detection, although urgently needed, remains elusive. Individuals in certain high-risk groups are, however, offered screening or surveillance. Here we explore advances in understanding high-risk groups for PDAC and efforts to implement biomarker-driven detection of PDAC in these groups. We review current approaches to early detection biomarker development and the use of artificial intelligence as applied to electronic health records (EHRs) and social media. Finally, we address the cost-effectiveness of applying biomarker strategies for early detection of PDAC.
Collapse
Affiliation(s)
- Kate Murray
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Lucy Oldfield
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Irena Stefanova
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | | | | | - Rachel O'Sullivan
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - William Greenhalf
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Salvatore Paiella
- Pancreatic Surgery Unit, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Italy
| | - Mateus N Aoki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Brazil
| | - Aldo Pastore
- Fondazione Pisana per la Scienza, Scuola Normale Superiore di Pisa, Italy
| | - James Birch-Ford
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Bhavana Hemantha Rao
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Czech Republic
| | - Pinar Uysal-Onganer
- School of Life Sciences, Cancer Mechanisms and Biomarkers Group, The University of Westminster, United Kingdom
| | - Caoimhe M Walsh
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - George B Hanna
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | | | | | | | | | - Andrei Turtoi
- Tumor Microenvironment and Resistance to Treatment Lab, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, France
| | - Elif Arik Sever
- Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | | | | | | | - Elif Öz
- Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | - Osman Uğur Sezerman
- Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | | | | | - Eithne Costello
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom.
| |
Collapse
|
2
|
Qi M, Zhang K, Zhang X, Zhu Y, Cai B, Wang C, Zhao G, Zhang D, Zhang J. Arginine tagged liposomal carrier for the delivery of celastrol for ferroptosis-induced hepatocellular carcinoma therapy. Colloids Surf B Biointerfaces 2025; 250:114546. [PMID: 39919344 DOI: 10.1016/j.colsurfb.2025.114546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/21/2025] [Accepted: 01/31/2025] [Indexed: 02/09/2025]
Abstract
Hepatocellular carcinoma (HCC) is a predominant malignant liver tumor that cannot be efficiently treated because of poor response, toxicity, and drug resistance. Ferroptosis is an iron-dependent way of cell death associated with abnormal intracellular lipid metabolism. Celastrol (Cel) has the ability to inhibit the progression of HCC by regulating multiple signaling pathways and induce ferroptosis. However, Cel exists the limitations of low water solubility, low oral bioavailability, and high organ toxicity. Cel was encapsulated in polyethylene glycol-based liposomes modified with L-arginine (Cel@Lip-Arg). Cel@Lip-Arg has a uniform size distribution (∼100 nm), high drug loading (80 %), and excellent ability to target liver cancer cells. In vitro experiments demonstrated that Cel@Lip-Arg considerably suppressed the activity of HuH7 (hepatoma) cells but had a negligible effect on L02 (normal) cells. Cel@Lip-Arg induced ferroptosis in hepatoma cells by promoting transferrin receptor expression, inhibiting system xc- and glutathione peroxidase 4, and favoring intracellular peroxide accumulation. In vivo experiments revealed that Cel@Lip-Arg plays a therapeutic role by inducing ferroptosis. Compared to Cel, Cel@Lip-Arg had a higher anti-hepatoma activity and effectively reduced the toxicity of Cel in mice. Cel@Lip-Arg-induced ferroptosis was concluded to be an attractive strategy for the precise treatment of HCC.
Collapse
Affiliation(s)
- Manman Qi
- School of Medicine, Shanghai University, Shanghai 200444, PR China
| | - Kai Zhang
- School of Medicine, Shanghai University, Shanghai 200444, PR China
| | - Xue Zhang
- School of Basic Medicine, Ningxia Medical University, Ningxia 750004, PR China
| | - Yuzhao Zhu
- Shanghai Universal Medical Imaging Diagnostic Center, Shanghai University, Shanghai 200233, PR China
| | - Banglan Cai
- School of Basic Medicine, Ningxia Medical University, Ningxia 750004, PR China
| | - Chao Wang
- Shanghai Universal Medical Imaging Diagnostic Center, Shanghai University, Shanghai 200233, PR China
| | - Gang Zhao
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Denghai Zhang
- School of Medicine, Shanghai University, Shanghai 200444, PR China.
| | - Jian Zhang
- School of Medicine, Shanghai University, Shanghai 200444, PR China; Shanghai Universal Medical Imaging Diagnostic Center, Shanghai University, Shanghai 200233, PR China.
| |
Collapse
|
3
|
Mansourabadi Z, Assarehzadegan MA, Mehdipour F, Ariafar A, Faghih Z, Safari E. Neutrophil extracellular traps and reactive oxygen species: Predictors of prognosis in bladder cancer. Immunol Lett 2025; 273:106991. [PMID: 39971199 DOI: 10.1016/j.imlet.2025.106991] [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: 07/15/2024] [Revised: 01/09/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
Neutrophils, the most abundant leukocytes in circulation, have become the subject of intensive research due to growing evidence of their role as modulators of cancer with both anti- and pro-tumorigenic effects. However, their prognostic function related to the release of neutrophil extracellular traps (NETs) and production of reactive oxygen species (ROS) has not yet been elucidated in the context of bladder cancer (BC). This study aimed to evaluate the ability of circulating neutrophils from BC patients to undergo NETosis and produce ROS-both spontaneously and following activation with phorbol 12-myristate 13-acetate (PMA)-using flow cytometry and immunofluorescence techniques. Their relevance to clinicopathological characteristics was also evaluated. Our results showed that PMA-treated neutrophils had increased early NETosis in patients with stage II (P = 0.048) and T2 (P = 0.014) compared to those with stage III and T3, respectively. These cells also showed a significant increase in ROS production in patients with T2 compared to those with T3 (P = 0.026) and T4 (P = 0.014), as well as in patients with stage II compared to stage IV (P = 0.048). Additionally, spontaneous ROS production was higher in patients without lymphovascular invasion than in those with invasion (P = 0.013). The increased activity of neutrophils observed in earlier stages (stage II and T2) suggests a potential protective mechanism in the early phases of cancer progression. It also highlights NETosis and ROS production by neutrophils as possible biomarkers for assessing disease progression. These findings provide insights into the complex interactions of neutrophils within the tumor microenvironment and lay the groundwork for further investigations into targeted therapies, potentially improving prognostic evaluations and treatment outcomes for patients.
Collapse
Affiliation(s)
- Zahra Mansourabadi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad-Ali Assarehzadegan
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Mehdipour
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Ariafar
- Department of Urology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Faghih
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Elahe Safari
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Breast Health and Cancer Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Shen J, Lin H, Mo K, Liang Z, Zhang Y, Quan H, Wang X, Zhang C, Chen C. Bidirectional roles of neutrophil extracellular traps in oral microbiota carcinogenesis: A systematic review. Transl Oncol 2025; 56:102361. [PMID: 40239243 PMCID: PMC12022684 DOI: 10.1016/j.tranon.2025.102361] [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: 08/30/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Neutrophil extracellular traps (NETs) are network structures composed of DNA, histones, and antimicrobial proteins,released by activated neutrophils to trap and eliminate extracellular pathogens. Recent research has demonstrated a strong correlation between NETs and various diseases, including immune dysregulation, thrombosis, and malignancies. This review synthesizes current research on NETs, focusing on its biological role in oral squamous cell carcinoma (OSCC) and explores its potential in treating. METHODS A literature review in the PubMed database was conducted to examine the impact of NETs on the homeostasis of oral microbiota and the involvement in the development of oral microbiota-related carcinogenesis. RESULTS Various microorganisms, including Porphyromonas gingivalis, Fusobacterium nucleatum, Streptococcus spp., along with Candida albicans, as well as certain viruses such as Human papillomavirus (HPV), Human herpes virus 8 (HHV-8), and Herpes simplex virus-1 (HSV-1)are regulated by NETs during oral colonization and proliferation and have been identified as contributors to the pathogenesis of oral squamous cell carcinoma. NETs have been shown to play a dual role in the carcinogenic process of oral microbiota in humans. At the initial stage of tumor formation, NETs inhibit tumorigenesis by eliminating tumorigenic bacteria that infiltrated the tumor; however, following tumor establishment, various cytokines and chemokines that promote tumor progression are released by neutrophils during the NETs formation. CONCLUSION This article reviews the oncogenic mechanisms of NETs in the oral microbiota, with potential implications for early tumor detection and the development of microbe-targeted therapies.
Collapse
Affiliation(s)
- Jie Shen
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Haitao Lin
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
| | - Kangnan Mo
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Zhong Liang
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yan Zhang
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Huatao Quan
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Xing Wang
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Chenping Zhang
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Chao Chen
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
5
|
Zhao X, Wang Y, Li J, Liu W, Yang Y, Qiao Y, Liao J, Chen M, Li D, Wu B, Huang D, Wu D. A machine-learning-derived online prediction model for depression risk in COPD patients: A retrospective cohort study from CHARLS. J Affect Disord 2025; 377:284-293. [PMID: 39988142 DOI: 10.1016/j.jad.2025.02.063] [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: 11/02/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Depression associated with Chronic Obstructive Pulmonary Disease (COPD) is a detrimental complication that significantly impairs patients' quality of life. This study aims to develop an online predictive model to estimate the risk of depression in COPD patients. METHODS This study included 2921 COPD patients from the 2018 China Health and Retirement Longitudinal Study (CHARLS), analyzing 36 behavioral, health, psychological, and socio-demographic indicators. LASSO regression filtered predictive factors, and six machine learning models-Logistic Regression, Support Vector Machine, Multilayer Perceptron, LightGBM, XGBoost, and Random Forest-were applied to identify the best model for predicting depression risk in COPD patients. Temporal validation used 2013 CHARLS data. We developed a personalized, interpretable risk prediction platform using SHAP. RESULTS A total of 2921 patients with COPD were included in the analysis, of whom 1451 (49.7 %) presented with depressive symptoms. 11 variables were selected to develop 6 machine learning models. Among these, the XGBoost model exhibited exceptional predictive performance in terms of discrimination, calibration, and clinical applicability, with an AUROC range of 0.747-0.811. In validation sets encompassing diverse population characteristics, XGBoost achieved the highest accuracy (70.63 %), sensitivity (59.05 %), and F1 score (63.17 %). LIMITATIONS The target population for the model is COPD patients. And the clinical benefits of interventions based on the prediction results remain uncertain. CONCLUSION We developed an online prediction platform for clinical application, allowing healthcare professionals to swiftly and efficiently evaluate the risk of depression in COPD patients, facilitating timely interventions and treatments.
Collapse
Affiliation(s)
- Xuanna Zhao
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524013, China
| | - Yunan Wang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524013, China
| | - Jiahua Li
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524013, China
| | - Weiliang Liu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524013, China
| | - Yuting Yang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524013, China
| | - Youping Qiao
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524013, China
| | - Jinyu Liao
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524013, China
| | - Min Chen
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524013, China
| | - Dongming Li
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524013, China
| | - Bin Wu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524013, China
| | - Dan Huang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524013, China
| | - Dong Wu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524013, China.
| |
Collapse
|
6
|
Novruzov F, Mehdi E, Aliyeva N, Orucova P, Simecek J, Aliyev J. The true negative [⁶⁸Ga]Ga-Trivehexin PET/CT in Solid Pseudopapillary Neoplasm of pancreas, mimicking pancreatic adenocarcinoma in [¹⁸F]FDG and [⁶⁸Ga]Ga-FAPI scans. Eur J Nucl Med Mol Imaging 2025; 52:1942-1943. [PMID: 39611956 DOI: 10.1007/s00259-024-06972-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/01/2024] [Indexed: 11/30/2024]
Affiliation(s)
- Fuad Novruzov
- Azerbaijan National Centre of Oncology, Department of Nuclear Medicine, M. Xiyabani street 137, AZ, 1011, Baku, Azerbaijan.
| | - Elnur Mehdi
- Azerbaijan National Centre of Oncology, Department of Nuclear Medicine, M. Xiyabani street 137, AZ, 1011, Baku, Azerbaijan
| | - Narmin Aliyeva
- Azerbaijan National Centre of Oncology, Department of Nuclear Medicine, M. Xiyabani street 137, AZ, 1011, Baku, Azerbaijan
| | - Parvin Orucova
- Azerbaijan National Centre of Oncology, Department of Pathology, Baku, Azerbaijan
| | | | - Jamil Aliyev
- Azerbaijan National Centre of Oncology, Department of General Surgery, Baku, Azerbaijan
| |
Collapse
|
7
|
Liu Y, Peng J, Zhao Y, Wang W. Emerging pathological diagnostic strategies for solid pseudopapillary neoplasm of the pancreas: insights from omics and innovative techniques. J Pathol Clin Res 2025; 11:e70029. [PMID: 40312910 PMCID: PMC12046068 DOI: 10.1002/2056-4538.70029] [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: 12/11/2024] [Revised: 02/21/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025]
Abstract
Solid pseudopapillary neoplasm (SPN) of the pancreas is a rare, low-grade malignant tumor, representing 0.9-2.7% of all exocrine pancreatic tumors. SPN patients generally have a favorable prognosis with a 5-year survival rate exceeding 95% following complete surgical resection. Accurate diagnosis is crucial to avoid unnecessary treatments. Currently, SPN diagnosis relies on imaging techniques such as CT and MRI, along with immunohistochemical analysis of biopsy and resection samples. The main challenge in diagnosis is the potential inability to accurately identify recurrent or metastatic SPN, as well as 'malignant' SPN, due to the lack of specific biomarkers. Advances in high-throughput omics technologies, including genomics, transcriptomics, proteomics and metabolomics, have opened new avenues for identifying novel biomarkers for SPN. Additional, liquid biopsy techniques have enabled more comprehensive analysis of biosamples such as pancreatic cyst fluid, offering promising prospects for preoperative diagnosis. This review highlights recent research on SPN diagnosis, focusing on immunohistochemical markers, tissue sampling methods and the potential of omics approaches. It also discusses the challenges and opportunities in improving diagnostic accuracy, particularly for high-grade and metastatic SPNs.
Collapse
Affiliation(s)
- Yuanhao Liu
- Department of PathologyPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
| | - Junya Peng
- Institute of Clinical MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
- State Key Laboratory of Complex, Severe, and Rare DiseasesPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
| | - Yupei Zhao
- State Key Laboratory of Complex, Severe, and Rare DiseasesPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
- Department of General SurgeryPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
- Department of Basic Medical SciencesSchool of Medicine, Tsinghua UniversityBeijingPR China
- Peking University‐Tsinghua Center for Life SciencesBeijingPR China
| | - Wenze Wang
- Department of PathologyPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
- Molecular Pathology Research Center, Department of PathologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPR China
| |
Collapse
|
8
|
Wang C, Tian H, Shang J. Exploration and perspectives on postoperative hyperlipasemia as a predictor of pancreatic fistula. Surgery 2025; 181:109025. [PMID: 39732550 DOI: 10.1016/j.surg.2024.109025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 11/30/2024] [Indexed: 12/30/2024]
Affiliation(s)
- Chenxi Wang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China.
| | - Huichuan Tian
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China
| | - Jin Shang
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
9
|
Ebrahim NAA, Othman MO, Salama RA, Abdelfatah D, Tahoun NS. Diagnostic insights into solid pseudopapillary neoplasms of the pancreas: a decade of experience with pediatric representation. Diagn Pathol 2025; 20:57. [PMID: 40307756 PMCID: PMC12042314 DOI: 10.1186/s13000-025-01648-9] [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: 12/27/2024] [Accepted: 04/11/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Solid pseudopapillary neoplasms (SPNs) of the pancreas are rare, low-grade malignancies that predominantly affect young females. Their diagnosis is often facilitated by a characteristic histomorphological pattern and immunohistochemical profile. However, diagnostic challenges persist, especially in pediatric and atypical presentations. Recent attention has focused on the diagnostic value of CD99 and LEF1 in distinguishing SPNs from other pancreatic neoplasms. OBJECTIVE To evaluate the diagnostic accuracy and utility of CD99 and LEF1 as immunohistochemical markers for SPNs. METHODS A retrospective analysis of 60 SPN cases diagnosed between 2015 and 2024 was performed. Histopathological features were systematically reviewed, and immunohistochemical staining for CD99, LEF1, β-catenin, Cyclin D1, PR, Ki-67 was evaluated. Immunohistochemical marker interpretation was standardized using internally validated thresholds informed by existing literature: CD99 was considered positive with ≥ 10% cytoplasmic staining exhibiting paranuclear accentuation; β-catenin positivity was defined by ≥ 5% nuclear localization; Cyclin D1 by ≥ 10% moderate-to-strong nuclear staining; and progesterone receptor (PR) expression by ≥ 1% nuclear positivity, consistent with hormone receptor evaluation guidelines. Marker expression was statistically analyzed for their associations. RESULTS SPNs exhibited a strong female predilection (F:M ratio ≈ 7:1), with a mean age of 32.5 years. Pediatric cases (n = 4) displayed higher mean expression of CD99 (73.8%) and LEF1 (86.5%) compared to adults. CD99 showed cytoplasmic positivity with paranuclear accentuation in 96.7% of cases, while LEF1 demonstrated nuclear staining in 91.7%. β-catenin nuclear localization was observed in 95% of tumors, reflecting Wnt/β-catenin pathway activation. Cyclin D1 and PR were expressed in 90% and 88.3% of cases, respectively. Co-expression of β-catenin, CD99, LEF1, Cyclin D1, and PR was observed in 73.3% of tumors. CD99 and LEF1 inversely correlated with tumor size and proliferative activity (Ki-67), whereas Cyclin D1 and Ki-67 positively correlated with tumor size and lymphovascular invasion (LVI). Pediatric tumors generally exhibited favorable profiles, with limited evidence of LVI. CONCLUSION SPNs present with distinctive immunohistochemical signatures that are critical for accurate diagnosis, particularly in morphologically ambiguous or pediatric cases. CD99 and LEF1 are highly sensitive markers that, in combination with β-catenin and Cyclin D1, enhance diagnostic precision. These findings emphasize the central role of Wnt/β-catenin signaling in SPN pathogenesis and underscore the importance of integrating clinicopathological and molecular data for comprehensive tumor assessment.
Collapse
Affiliation(s)
- Noura A A Ebrahim
- Oncologic Pathology Department, National Cancer Institute (NCI) - Cairo University, Cairo, Egypt.
| | - Moamen O Othman
- Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Rasha A Salama
- Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE
| | - Dalia Abdelfatah
- Cancer Epidemiology and Biostatistics Department, National Cancer Institute (NCI) - Cairo University, Cairo, Egypt
| | - Neveen S Tahoun
- Oncologic Pathology Department, National Cancer Institute (NCI) - Cairo University, Cairo, Egypt
| |
Collapse
|
10
|
Rafiepoor H, Banoei MM, Ghorbankhanloo A, Muhammadnejad A, Razavirad A, Soleymanjahi S, Amanpour S. Exploring the potential of machine learning in gastric cancer: prognostic biomarkers, subtyping, and stratification. BMC Cancer 2025; 25:809. [PMID: 40307780 PMCID: PMC12042310 DOI: 10.1186/s12885-025-14204-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: 12/29/2024] [Accepted: 04/23/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Advancements in the management of gastric cancer (GC) and innovative therapeutic approaches highlight the significance of the role of biomarkers in GC prognosis. Machine-learning (ML)-based methods can be applied to identify the most important predictors and unravel their interactions to classify patients, which might guide prioritized treatment decisions. METHODS A total of 140 patients with histopathological confirmed GC who underwent surgery between 2011 and 2016 were enrolled in the study. The inspired modification of the partial least squares (SIMPLS)-based model was used to identify the most significant predictors and interactions between variables. Predictive partition analysis was employed to establish the decision tree model to prioritize markers for clinical use. ML models have also been developed to predict TNM stage and different subtypes of GC. Latent class analysis (LCA) and principal component analysis (PCA) were carried out to cluster the GC patients and to find a subgroup of survivors who tended to die. RESULTS The findings revealed that the SIMPLS method was able to predict the mortality of GC patients with high predictabilities (Q2 = 0.45-0.70). The analysis identified MMP-7, P53, Ki67, and vimentin as the top predictors. Correlation analysis revealed different patterns of prognostic markers in the non-survivor and survivor cohorts and different GC subtypes. The main prediction models were verified via other ML-based analyses, with a high area under the curve (AUC) (0.84-0.99), specificity (0.82-0.99) and sensitivity (0.87-0.99). Patients were classified into three clusters of mortality risk, which highlighted the most significant mortality predictors. Partition analysis prioritizes the most significant predictors P53 ≥ 6, COX-2 > 2, vimentin > 2, Ki67 ≥ 13 in mortality of patients (AUC = 0.85-0.90). CONCLUSION The present study highlights the importance of considering multiple variables and their interactions to predict the prognosis of mortality and stage in GC patients through ML-based techniques. These findings suggest that the incorporation of molecular biomarkers may enhance patient prognosis compared to relying solely on clinical factors. Furthermore, they demonstrate the potential for personalized medicine in GC treatment by identifying high-risk patients for early intervention and optimizing therapeutic strategies. The partition analysis technique offers a practical tool for identifying cutoffs and prioritizing markers for clinical application. Additionally, providing Clinical Decision Support systems with predictive tools can assist clinicians and pathologists in identifying aggressive cases, thereby improving patient outcomes while minimizing unnecessary treatments. Overall, this study contributes to the ongoing efforts to improve patient outcomes by advancing our comprehension of the intricate nature of GC.
Collapse
Affiliation(s)
- Haniyeh Rafiepoor
- Cancer Biology Research Center, Cancer Institute, Tehran University of Medical Sciences, Keshavarz Blvd, Building, Tehran, Iran
| | - Mohammad M Banoei
- Department of Critical Care Medicine, University of Calgary, Calgary, AB, Canada
- Department of Biological Science, University of Calgary, Calgary, AB, Canada
| | - Alireza Ghorbankhanloo
- Cancer Biology Research Center, Cancer Institute, Tehran University of Medical Sciences, Keshavarz Blvd, Building, Tehran, Iran
| | - Ahad Muhammadnejad
- Cancer Biology Research Center, Cancer Institute, Tehran University of Medical Sciences, Keshavarz Blvd, Building, Tehran, Iran
| | - Amirhossein Razavirad
- Cancer Biology Research Center, Cancer Institute, Tehran University of Medical Sciences, Keshavarz Blvd, Building, Tehran, Iran
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Soleymanjahi
- Department of Internal Medicine, Department of Digital Health, Yale School of Medicine, New Haven, CT, USA
| | - Saeid Amanpour
- Cancer Biology Research Center, Cancer Institute, Tehran University of Medical Sciences, Keshavarz Blvd, Building, Tehran, Iran.
| |
Collapse
|
11
|
Wang Q, Wang Z, Liu F, Wang Z, Ni Q, Chang H. Machine learning-based prediction of postoperative pancreatic fistula after laparoscopic pancreaticoduodenectomy. BMC Surg 2025; 25:191. [PMID: 40307833 PMCID: PMC12045007 DOI: 10.1186/s12893-025-02935-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Clinically relevant postoperative pancreatic fistula (CR-POPF) following laparoscopic pancreaticoduodenectomy (LPD) is a critical complication that significantly worsens patient outcomes. However, the heterogeneity of its risk factors and the clinical utility of predictive models remain to be fully elucidated. This study aims to systematically analyze the risk factors for CR-POPF and develop an optimized predictive model using machine learning algorithms, providing an evidence-based approach for individualized risk assessment in patients undergoing LPD. METHODS A retrospective study was conducted, including 210 patients with periampullary cancer who underwent laparoscopic pancreaticoduodenectomy (LPD) at the Hepatobiliary Surgery Center, Olympic Stadium Campus, Shandong Provincial Hospital Affiliated to Shandong First Medical University, from January 2017 to January 2024. Patients were classified into the clinically relevant pancreatic fistula (CR-POPF) group (n = 34) and the non-clinically relevant pancreatic fistula (non-CR-POPF) group (n = 176) according to the 2016 criteria of the International Study Group of Pancreatic Surgery (ISGPS). Potential risk factors were identified through intergroup comparisons, and independent risk factors were determined using univariate and multivariate logistic regression analyses. Based on these findings, a predictive model for CR-POPF was developed using machine learning algorithms. RESULTS CR-POPF was associated with higher BMI, monocyte levels, platelet count, total bilirubin, AST, ALT, and lower albumin. Pathological diagnosis of ampullary carcinoma and soft pancreatic texture were significantly more common in the CR-POPF group. Multivariate analysis identified soft pancreatic texture as an independent predictor (OR = 4.99, 95% CI: 1.93-12.86). Among all models, the random forest model showed the best performance (AUC = 0.747, sensitivity = 0.917, specificity = 0.574), using only preoperative variables such as age, gender, BMI, hypertension, diabetes, hemoglobin, platelets, AST, and ALT. CONCLUSION Soft pancreatic texture was identified as an independent risk factor for postoperative pancreatic fistula following laparoscopic pancreaticoduodenectomy (LPD). The random forest model based on preoperative clinical variables enables individualized risk prediction, offering value for preoperative planning and postoperative care.
Collapse
Affiliation(s)
- Qianchang Wang
- Shandong First Medical University, Jinan, Shandong, China
| | - Zhe Wang
- Shandong First Medical University, Jinan, Shandong, China
| | - Fangfeng Liu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University: Shandong Provincial Hospital, Jinan, Shandong, China.
| | - Zhengjian Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University: Shandong Provincial Hospital, Jinan, Shandong, China
| | - Qingqiang Ni
- Shandong Provincial Hospital Affiliated to Shandong First Medical University: Shandong Provincial Hospital, Jinan, Shandong, China
| | - Hong Chang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University: Shandong Provincial Hospital, Jinan, Shandong, China
| |
Collapse
|
12
|
Li L, Zeng Y, Cheng G, Yang H. Acetylation and deacetylation dynamics in stress response to cancer and infections. Semin Immunol 2025; 78:101957. [PMID: 40288003 DOI: 10.1016/j.smim.2025.101957] [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: 05/08/2024] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
In response to stress stimuli, cells have evolved various mechanisms to integrate internal and external signals to achieve dynamic homeostasis. Lysine acetyltransferase (KATs) and deacetyltransferase (KDACs) are the key modulators of epigenetic modifications, enabling cells to modulate cellular responses through the acetylation and deacetylation of both histone and nonhistone proteins. Understanding the signaling pathways involved in cellular stress response, along with the roles of KATs and KDACs may pave the way for the development of novel therapeutic strategies. This review discusses the molecular mechanisms of acetylation and deacetylation in stress responses related to tumorigenesis, viral and bacterial infections. In tumorigenesis section, we focused on the tumor cells' intrinsic and external molecules and signaling pathways regulated by acetylation and deacetylation modification. In viral and bacterial infections, we summarized the update research on acetylation and deacetylation modification in viral and bacterial infections, which systematical introduction on this topic is not too much. Additionally, we provide an overview of current therapeutic interventions and clinical trials involving KAT and KDAC inhibitors in the treatment of cancer, as well as viral and bacterial infection-related diseases.
Collapse
Affiliation(s)
- Lili Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Guangzhou National Laboratory, Guangzhou, Guangdong 510005, China
| | - Yanqiong Zeng
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China
| | - Genhong Cheng
- Guangzhou National Laboratory, Guangzhou, Guangdong 510005, China
| | - Heng Yang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China
| |
Collapse
|
13
|
Kaiser JD, Bräuherr F, Biesel EA, Chikhladze S, Fichtner-Feigl S, Ruess DA, Wittel UA. Preoperative prediction of postoperative pancreatic fistula after Pancreaticoduodenectomy: Determination and validation of a cut-off value for the Roberts Score. Am J Surg 2025; 245:116356. [PMID: 40319558 DOI: 10.1016/j.amjsurg.2025.116356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND POPF after pancreaticoduodenectomy can be life-threatening. For risk stratification, prediction could be key. The aim of this study is to determine and validate a cut-off value for the Roberts Score, which is one of the few purely preoperative multicenter validated predictive models for POPF. METHODS 582 patients were included. The Youden index determined a cut-off in the exploratory cohort (n = 466). The validation cohort's (n = 116) ability to predict CR-POPF was tested using univariate and multivariate regression analysis. RESULTS AUC of Roberts Score for the exploration cohort was 0.768. The identified cut-off of 0.268 was confirmed in the validation cohort (p < 0.001). Higher scores were significantly associated with longer time to drain removal and ICU stay. Multiple logistic regression showed the cut-off as an independent predictor of CR-POPF (p = 0.038). CONCLUSION The scoring variables and the cut-off itself were both independent predictors, which may improve the identification of high-risk patients and help to investigate the development of POPF.
Collapse
Affiliation(s)
- Johannes D Kaiser
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany.
| | - Franziska Bräuherr
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Esther A Biesel
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Sophia Chikhladze
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Stefan Fichtner-Feigl
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Dietrich A Ruess
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Uwe A Wittel
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
14
|
Yi S, Noh K, Kim H, Jung E, Kim S, Lee J, Guk K, Choi J, Lim EK, Kim S, Park H, Lim JH, Jung CR, Kang T, Jung J. Advancing pancreatic cancer therapy by mesothelin-specific nanobody conjugation. Mol Cancer 2025; 24:124. [PMID: 40275270 PMCID: PMC12020020 DOI: 10.1186/s12943-025-02325-7] [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: 11/20/2024] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is highly challenging to treat due to its poor prognosis and limited effective treatment options. Liposomal nanotechnology has emerged as a promising drug delivery platform in oncology, but existing liposomal therapies face limitations such as systemic toxicity, insufficient tumor selectivity, and low target specificity. Mesothelin (MSLN), an antigen overexpressed in PAAD, has attracted attention as a potential target for precision therapy. Here, we present the development of an anti-MSLN nanobody (D3 Nb) with high binding affinity (KD = 2.2 nM) that can selectively bind to MSLN-positive cancer cells. Structural analysis revealed that hydrophobic and hydrogen bonds within the complementary determining region (CDR) of D3 Nb promote strong binding to MSLN, leading to significant inhibition of AKT/NF-κB signaling and downregulation of fibronectin 1 (FN1) and twist1, key drivers of PAAD oncogenicity. In vivo studies confirmed that D3 Nb alone inhibits tumor progression. Furthermore, selective delivery to MSLN-positive tumors in combination with gemcitabine-loaded liposomes (D3-LNP-GEM) significantly improved cytotoxicity and promoted tumor regression. These findings highlight the potential of the D3-LNP-GEM platform as a novel targeted therapy for MSLN-expressing malignancies, showing promising efficacy in preclinical models and paving the way for continued clinical evaluation.
Collapse
Affiliation(s)
- Soyeon Yi
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Kyunghee Noh
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
- Department of Nanobiotechnology, KRIBB School of Biotechnology, UST, 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Hyeran Kim
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Eunkyeong Jung
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Suhyeon Kim
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
- Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, 26 Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Republic of Korea
| | - Jieun Lee
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Kyeonghye Guk
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jinsol Choi
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Eun-Kyung Lim
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
- Department of Nanobiotechnology, KRIBB School of Biotechnology, UST, 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, 16419, Republic of Korea
| | - Seokho Kim
- Department of Health Science, the Graduate School of Dong-A University, Busan, 49315, Republic of Korea
| | - Hwangseo Park
- Deparment of Bioscience and Biotechnology, Sejong University, 209 Neungdong-ro, Kwangjin- gu, Seoul, 05006, Republic of Korea
| | - Jung Hwa Lim
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, Republic of Korea
- Department of Functional Genomics, Korea University of Science and Technology (UST), 217 Gajeong-ro, Daejeon, Republic of Korea
| | - Cho-Rok Jung
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, Republic of Korea
- Department of Functional Genomics, Korea University of Science and Technology (UST), 217 Gajeong-ro, Daejeon, Republic of Korea
| | - Taejoon Kang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, 16419, Republic of Korea
| | - Juyeon Jung
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
- Department of Nanobiotechnology, KRIBB School of Biotechnology, UST, 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea.
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, 16419, Republic of Korea.
| |
Collapse
|
15
|
Michetti F, Cirone M, Strippoli R, D'Orazi G, Cordani M. Mechanistic insights and therapeutic strategies for targeting autophagy in pancreatic ductal adenocarcinoma. Discov Oncol 2025; 16:592. [PMID: 40266451 PMCID: PMC12018664 DOI: 10.1007/s12672-025-02400-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/15/2025] [Indexed: 04/24/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterised by early metastasis and resistance to anti-cancer therapy, leading to an overall poor prognosis. Macroautophagy (hereinafter referred to as autophagy) is a conserved cellular homeostasis mechanism that degrades various cargoes (e.g., proteins, organelles, and pathogens) mainly playing a role in promoting survival under environmental stress. Autophagy is an essential defense mechanism against PDAC initiation, acting on multiple levels to maintain cellular and tissue homeostasis. However, autophagy is also intimately involved in the molecular mechanisms driving PDAC progression, facilitating the adaptation of cancer cells to the tumor microenvironment's harsh conditions. In this review, we examine the complex role of autophagy in PDAC and assess the potential of modulating autophagy as a therapeutic strategy. By reviewing current research and clinical trials, we seek to elucidate how targeting autophagy can disrupt PDAC tumor survival mechanisms, enhance the efficacy of existing treatments, and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Federica Michetti
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149, Rome, Italy
| | - Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149, Rome, Italy.
| | - Gabriella D'Orazi
- UniCamillus-Saint Camillus International University of Health and Medical Sciences, Via di Sant'Alessandro 8, 00131, Rome, Italy.
- Department of Research and Advanced Technologies, Regina Elena National Cancer Institute IRCCS, Via Elio Chianesi 51, 00144, Rome, Italy.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040, Madrid, Spain.
| |
Collapse
|
16
|
Ahmad GV, Nouri S, Mohammad Gholian A, Abdollahi E, Ghorbaninezhad F, Tahmasebi S, Eterafi M, Askari MR, Safarzadeh E. Breaking barriers: CAR-NK cell therapy breakthroughs in female-related cancers. Biomed Pharmacother 2025; 187:118071. [PMID: 40253831 DOI: 10.1016/j.biopha.2025.118071] [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: 01/16/2025] [Revised: 04/04/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025] Open
Abstract
Cancer stands as a leading cause of mortality globally. The main female-related malignancies are breast cancer, with 2.3 million new cases annually, and ovarian cancer, with 300,000 new cases per year worldwide. The current treatments like surgery, chemotherapy, and radiation therapy have presumably had deficiencies in sustaining long-term anti-tumor responses. Cellular immunotherapy, also referred to as adoptive cell therapy, has shown encouraging advances by employing genetically modified immune cells in fighting cancer by engineering chimeric antigen receptors (CARs) mainly on T cells and natural killer (NK) cells. Studies in NK cell therapies involve unmodified NK cells and CAR-NK cell therapies, targeting cancer cells while limiting the destruction of normal cells. CAR-NK cells represent the next generation of therapeutic immune cells that have been shown to eliminate malignancies through CAR-dependent and CAR-independent mechanisms. They also represent possible candidates for "off-the-shelf" therapies due to their advantages, including the ability to target cancer cells independently of the major histocompatibility complex, reduced risk of alloreactivity, and fewer severe toxicities compared to CAR-T cells. To date, there have been no comprehensive review studies examining the therapeutic potential of CAR-NK cell therapy specifically for female-related malignancies, such as breast and ovarian cancers. This review offers a thorough exploration of CAR-NK cell therapy in relation to these cancers and their responses to treatment.
Collapse
Affiliation(s)
- Ghorbani Vanan Ahmad
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Samaneh Nouri
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Eileen Abdollahi
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Farid Ghorbaninezhad
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Safa Tahmasebi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Majid Eterafi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Reza Askari
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Department of Microbiology, Parasitology, and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
17
|
Xia H, Lin J, Liu M, Lai J, Yang Z, Qiu L. Association of blood urea nitrogen to albumin ratio with mortality in acute pancreatitis. Sci Rep 2025; 15:13327. [PMID: 40247063 PMCID: PMC12006543 DOI: 10.1038/s41598-025-97891-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
Blood urea nitrogen (BUN) and serum albumin (ALB) are strongly associated with the prognosis in acute pancreatitis (AP). The BUN/ALB ratio (BAR) reflects renal, nutritional, inflammatory, and endothelial functions. In this study, we investigated the association between the BAR and all-cause mortality in critically ill patients with AP. Using data from the Medical Information Market for Intensive Care (MIMIC-IV) database, we conducted a retrospective cohort analysis. The relationship between BAR and mortality was assessed through Kaplan-Meier survival curves, restricted cubic spline models, and multivariable Cox proportional hazards regression. The predictive capacity of BAR for 30-day and 1-year mortality was evaluated using receiver operating characteristic analysis. Our study included 780 participants, with 30-day and 1-year mortality rates of 12.6% and 23.6%, respectively. Higher BAR values were associated with poorer survival outcomes. BAR demonstrated superior predictive performance achieving an area under the curve of 0.74, surpassing BUN, ALB, and SOFA scores. The Cox model indicated a significant independent association between elevated BAR and increased mortality risk, with hazard ratios of 1.43 (95% CI 1.20-1.70) for 30-day mortality and 1.37 (95% CI 1.17-1.60) for 1-year mortality. Stratified and sensitivity analyses confirmed the robustness of these findings. Our results suggest that elevated BAR is associated with poor prognosis in critically ill patients with AP and may serve as a valuable tool for early risk stratification and for assessing both short- and long-term prognosis.
Collapse
Affiliation(s)
- Hao Xia
- Department of Medicine Intensive Care Unit, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou, 363000, Fujian, China
| | - Jinzhan Lin
- Department of Medicine Intensive Care Unit, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou, 363000, Fujian, China
| | - Minchao Liu
- Department of Hepatobiliary Surgery, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou, 363000, Fujian, China
| | - Jiawei Lai
- Department of Medicine Intensive Care Unit, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou, 363000, Fujian, China
| | - Zhaobin Yang
- Department of Medicine Intensive Care Unit, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou, 363000, Fujian, China.
| | - Luzhen Qiu
- Department of Medicine Intensive Care Unit, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou, 363000, Fujian, China.
| |
Collapse
|
18
|
Li X, Lu N, Sun K, Shi F, Lin L, Chen Y, Wang Y, Wang M, Sun K, Xue X, Xiao W, Su X, Bai X, Liang T. [ 18F]FAPI- 04 PET/CT for pathologic response assessment in pancreatic cancer patients with systemic treatment. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07271-6. [PMID: 40237796 DOI: 10.1007/s00259-025-07271-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025]
Abstract
PURPOSE To study the association between [18F]FAPI- 04 uptake on positron emission tomography/computed tomography (PET/CT) and pathologic treatment response (PTR) in patients with pancreatic cancer (PC). METHODS We enrolled 59 patients from August 2021, of whom 28 underwent surgical treatment after systemic therapy. The patients were investigated for a correlation between baseline fibroblast activation protein inhibitor (FAPI) uptake and PTR using College of American Pathologists (CAP) scores. The FAPI PET variables include standardised uptake value (SUV)max, SUVmean, metabolic tumour volume (MTV), and total lesion FAP expression (TLF). A PET/CT scan obtained before surgery in 14 patients facilitated assessing changes in FAPI uptake through treatment, and their association with PTR. Multiplex immunohistochemistry (mIHC) analysis identified the FAPI biodistribution in the PC tumours. RESULTS The SUVmax correlated positively with FAP expression in PC tissues. However, there was no correlation between baseline variables and the CAP scores. Treatment resulted in remarkably reduced MTV and TLF in all patients. The baseline SUVmax and SUVmean of patients with a good PTR (CAP score ≤ 2) differed from those after treatment (p = 0.001). An FITC-FAPI probe intuitively showed that cancer-associated fibroblasts (CAFs) and tumour cells had a similar FITC-FAPI fluorescence intensity, indicating a negative association between tumour regression and [18F]FAPI- 04 uptake. CONCLUSION Greater changes in FAPI uptake through treatment were associated with a better PTR in patients with PC and might be valuable in predicting prolonged survival. These results are clinically meaningful when selecting candidates for conversion surgery during systemic treatment.
Collapse
Affiliation(s)
- Xiang Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
- Zhejiang University Cancer Center, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Na Lu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
- Zhejiang University Cancer Center, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Kang Sun
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
- Zhejiang University Cancer Center, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Fukang Shi
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
- Zhejiang University Cancer Center, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Lili Lin
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiwen Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
- Zhejiang University Cancer Center, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Yangyang Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
- Zhejiang University Cancer Center, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Meng Wang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke Sun
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xing Xue
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenbo Xiao
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinhui Su
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China.
- Zhejiang University Cancer Center, Hangzhou, China.
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China.
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China.
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China.
- Zhejiang University Cancer Center, Hangzhou, China.
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China.
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
19
|
Potievskiy MB, Petrov LO, Ivanov SA, Sokolov PV, Trifanov VS, Grishin NA, Moshurov RI, Shegai PV, Kaprin AD. Machine learning for modeling and identifying risk factors of pancreatic fistula. World J Gastrointest Oncol 2025; 17:100089. [PMID: 40235910 PMCID: PMC11995311 DOI: 10.4251/wjgo.v17.i4.100089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/05/2024] [Accepted: 02/05/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Pancreatic fistula is the most common complication of pancreatic surgeries that causes more serious conditions, including bleeding due to visceral vessel erosion and peritonitis. AIM To develop a machine learning (ML) model for postoperative pancreatic fistula and identify significant risk factors of the complication. METHODS A single-center retrospective clinical study was conducted which included 150 patients, who underwent pancreatoduodenectomy. Logistic regression, random forest, and CatBoost were employed for modeling the biochemical leak (symptomless fistula) and fistula grade B/C (clinically significant complication). The performance was estimated by receiver operating characteristic (ROC) area under the curve (AUC) after 5-fold cross-validation (20% testing and 80% training data). The risk factors were evaluated with the most accurate algorithm, based on the parameter "Importance" (Im), and Kendall correlation, P < 0.05. RESULTS The CatBoost algorithm was the most accurate with an AUC of 74%-86%. The study provided results of ML-based modeling and algorithm selection for pancreatic fistula prediction and risk factor evaluation. From 14 parameters we selected the main pre- and intraoperative prognostic factors of all the fistulas: Tumor vascular invasion (Im = 24.8%), age (Im = 18.6%), and body mass index (Im = 16.4%), AUC = 74%. The ML model showed that biochemical leak, blood and drain amylase level (Im = 21.6% and 16.4%), and blood leukocytes (Im = 11.2%) were crucial predictors for subsequent fistula B/C, AUC = 86%. Surgical techniques, morphology, and pancreatic duct diameter less than 3 mm were insignificant (Im < 5% and no correlations detected). The results were confirmed by correlation analysis. CONCLUSION This study highlights the key predictors of postoperative pancreatic fistula and establishes a robust ML-based model for individualized risk prediction. These findings contribute to the advancement of personalized perioperative care and may guide targeted preventive strategies.
Collapse
Affiliation(s)
- Mikhail B Potievskiy
- Center for Clinical Trials, Center for Innovative Radiological and Regenerative Technologies, FSBI “National Medical Research Radiological Center” of the Ministry of Health of the Russian Federation, Obninsk 249036, Kaluzhskaya Oblast, Russia
| | - Leonid O Petrov
- Department of Radiation and Surgical Treatment of Abdominal Diseases, A. Tsyb Medical Radiological Center, FSBI “National Medical Research Radiological Center” of the Ministry of Health of the Russian Federation, Obninsk 249036, Kaluzhskaya Oblast, Russia
| | - Sergei A Ivanov
- Department of Administration, FSBI “National Medical Research Radiological Center” of the Ministry of Health of the Russian Federation, Obninsk 249036, Kaluzhskaya Oblast, Russia
| | - Pavel V Sokolov
- Department of Operation Unit, FSBI “National Medical Research Radiological Center” of the Ministry of Health of the Russian Federation, Obninsk 249036, Kaluzhskaya Oblast, Russia
| | - Vladimir S Trifanov
- Department of Abdominal Oncology, P. Herzen Moscow Oncological Institute, FSBI “National Medical Research Radiological Center” of the Ministry of Health of the Russian Federation, Obninsk 249036, Kaluzhskaya Oblast, Russia
| | - Nikolai A Grishin
- Department of Abdominal Oncology, P. Herzen Moscow Oncological Institute, FSBI “National Medical Research Radiological Center” of the Ministry of Health of the Russian Federation, Obninsk 249036, Kaluzhskaya Oblast, Russia
| | - Ruslan I Moshurov
- Department of Abdominal Oncology, P. Herzen Moscow Oncological Institute, FSBI “National Medical Research Radiological Center” of the Ministry of Health of the Russian Federation, Obninsk 249036, Kaluzhskaya Oblast, Russia
| | - Peter V Shegai
- Center for Innovative Radiological and Regenerative Technologies, FSBI “National Medical Research Radiological Center” of the Ministry of Health of the Russian Federation, Obninsk 249036, Kaluzhskaya Oblast, Russia
| | - Andrei D Kaprin
- Department of Administration, FSBI “National Medical Research Radiological Center” of the Ministry of Health of the Russian Federation, Obninsk 249036, Kaluzhskaya Oblast, Russia
- Department of Urology and Operative Nephrology with Course of Oncology, Medical Faculty, Medical Institute, Peoples’ Friendship University of Russia, Moscow 117198, Moskva, Russia
| |
Collapse
|
20
|
Das C, Sepay N, Kim TW, Chae S, Ghosh N, Dumpala M, Choi D, Jeon S, Im J, Biswas G. Recycling Motorcycle Exhaust Soot into Fluorescent Graphene Oxide Quantum Dots for Sensing Ferrocyanide Ions and Bioimaging Cells: A Method for Waste Utilization. ACS OMEGA 2025; 10:14229-14240. [PMID: 40256545 PMCID: PMC12004190 DOI: 10.1021/acsomega.5c00229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025]
Abstract
Graphene oxide quantum dots (GOQDs) with a high quantum yield (50%) were synthesized using soot collected from a motorcycle (petroleum vehicle) exhaust pipe and applied as sensors for ferrocyanide ([Fe(CN)6]4-) ions and as bioimaging agents in a cancer cell line. X-ray photoelectron spectroscopy (XPS) data for the GOQDs revealed a C/O ratio of 2.49, which was close to that of graphene oxide (GO). The synthesized GOQDs exhibited strong blue fluorescence. High sensitivity to detect [Fe(CN)6]4- was reported in GOQDs with a detection limit of 0.46 nmol mL-1, and a strong linear relationship was achieved in the concentration range of 100-1100 μg L-1. The results demonstrate the utility of GOQDs for detecting [Fe(CN)6]4- in a real scenario. The GOQDs exhibited almost negligible cytotoxicity in cells and were internalized within 4 h of incubation, emitting blue fluorescence in the cytoplasm. This suggests that the GOQDs are promising bioimaging agents for biomedical applications. In general, these waste-derived GOQDs appear to be good chemo- and biosensing probes for real-life applications.
Collapse
Affiliation(s)
- Chanchal Das
- Department
of Chemistry, Cooch Behar Panchanan Barma
University, Cooch
Behar 736101, India
| | - Nasim Sepay
- Department
of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, Asan 31538, Republic
of Korea
| | - Tae Wan Kim
- Department
of Medical Life Science, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Shinwon Chae
- Department
of Biochemistry, Soonchunhyang University,
College of Medicine, Cheonan 31151, Republic of Korea
| | - Nandan Ghosh
- Department
of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, Asan 31538, Republic
of Korea
| | - Mohan Dumpala
- Department
of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, Asan 31538, Republic
of Korea
| | - Dongsic Choi
- Department
of Biochemistry, Soonchunhyang University,
College of Medicine, Cheonan 31151, Republic of Korea
| | - Seob Jeon
- Department
of Obstetrics and Gynecology, College of
Medicine, Soonchunhyang University Cheonan Hospital, Cheonan 31151, Republic of Korea
| | - Jungkyun Im
- Department
of Electronic Materials, Devices, and Equipment Engineering, Soonchunhyang University, Asan 31538, Republic
of Korea
- Department
of Chemical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Goutam Biswas
- Department
of Chemistry, Cooch Behar Panchanan Barma
University, Cooch
Behar 736101, India
| |
Collapse
|
21
|
Sekiguchi N, Takahashi H, Kobayashi S, Sasaki K, Hasegawa S, Iwagami Y, Yamada D, Tomimaru Y, Akita H, Asaoka T, Noda T, Shimizu J, Doki Y, Eguchi H. Surgical outcomes of histopathological stage I pancreatic cancer with special reference to oncological differences between pStage I and ypStage I cases. Surg Oncol 2025; 60:102225. [PMID: 40252582 DOI: 10.1016/j.suronc.2025.102225] [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: 01/18/2025] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 04/21/2025]
Abstract
OBJECTIVES Histopathological stage I pancreatic cancer (PC) consists of four entities according to the Union for International Cancer Control (UICC) 8th staging system: pStage IA (cases underwent up-front surgery, pT1 [tumor size ≤20 mm] pN0), pStage IB (cases underwent up-front surgery, pT2 [20 mm < tumor size ≤40 mm] pN0), ypStage IA and IB (i.e., underwent neoadjuvant treatment [NAT]). This study aimed to evaluate surgical outcomes in patients with histopathological stage I PC ((y)pStage I). METHODS Patients with PC who underwent resection and were diagnosed with (y)pStage I were included in this study (n = 121). We evaluated and compared recurrence-free survival (RFS) and overall survival (OS) among these four categories: pStage I vs. ypStage I, (y)pStage IA vs. IB, ypStage IA vs. IB, and pStage IA vs. IB. RESULTS There was no difference in the prognosis between pStage I and ypStage I cases, even though ypStage I included more advanced cases at NAT initiation. For pStage I (n = 25), no differences in prognosis were noted between pStage IA and IB (median RFS, 24 vs. 21 months, p = 0.871; median OS, 49.5 vs. 35.5 months, p = 0.213). However, in the ypStage I cohort (n = 96), there was significantly better RFS in ypStage IA (median RFS, 42 vs. 14 months, p = 0.0114; median OS, 52 vs. 36 months, p = 0.250). CONCLUSIONS Survival was comparable between pStage I and ypStage I cohorts, although the subcategories within each cohort (IA vs. IB) had different clinicopathological characteristics and outcomes.
Collapse
Affiliation(s)
- Naoko Sekiguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2 E2, Suita, Osaka, 565-0871, Japan
| | - Hidenori Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2 E2, Suita, Osaka, 565-0871, Japan.
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2 E2, Suita, Osaka, 565-0871, Japan
| | - Kazuki Sasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2 E2, Suita, Osaka, 565-0871, Japan
| | - Shinichiro Hasegawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2 E2, Suita, Osaka, 565-0871, Japan
| | - Yoshifumi Iwagami
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2 E2, Suita, Osaka, 565-0871, Japan
| | - Daisaku Yamada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2 E2, Suita, Osaka, 565-0871, Japan
| | - Yoshito Tomimaru
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2 E2, Suita, Osaka, 565-0871, Japan
| | - Hirofumi Akita
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2 E2, Suita, Osaka, 565-0871, Japan
| | - Tadafumi Asaoka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2 E2, Suita, Osaka, 565-0871, Japan
| | - Takehiro Noda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2 E2, Suita, Osaka, 565-0871, Japan
| | - Junzo Shimizu
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2 E2, Suita, Osaka, 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2 E2, Suita, Osaka, 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2 E2, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
22
|
Yu H, Jiang Y, Miao W, Hu W, Jin Y, Fan Z, Luo P, Tao R, Zhu F, Han R, Zhou J. The trend in pancreatic cancer incidence from 2009 to 2019 and the prediction from 2020 to 2030: An analysis of provincial data in China. Public Health 2025; 243:105693. [PMID: 40222142 DOI: 10.1016/j.puhe.2025.03.007] [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: 05/08/2024] [Revised: 02/09/2025] [Accepted: 03/11/2025] [Indexed: 04/15/2025]
Abstract
OBJECTIVE As a malignant tumour with a very high mortality rate, the incidence of pancreatic cancer is on the rise globally, but the long-term trend at the provincial level in China is not yet clear. This study aimed to investigate the trend of pancreatic cancer incidence in Jiangsu Province from 2009 to 2019 and to predict pancreatic cancer incidence from 2020 to 2030. STUDY DESIGN Descriptive study. METHODS Data on pancreatic cancer incidence in Jiangsu Province were obtained from the Jiangsu Cancer Registry. Trends in pancreatic cancer incidence from 2009 to 2019 were examined based on the Joinpoint regression model. Age-period-cohort (APC) models were introduced to estimate the independent effects of age, period, and cohort on the incidence of pancreatic cancer and to project pancreatic cancer incidence from 2020 to 2030. RESULTS From 2009 to 2019, the number of pancreatic cancer cases in Jiangsu Province increased from 1146 to 2088, and the age-standardized incidence rate (ASIR) increased from 4.59 to 5.64 per 100,000 people. In 2019, the ASIR was higher in males than in females, and the ASIR was higher in urban areas than in rural areas. The APC analysis also showed that the age effect was the most important factor influencing pancreatic cancer incidence in Jiangsu Province. Predictions suggest that pancreatic cancer incidence will continue to increase from 2020 to 2030. CONCLUSIONS From 2009 to 2019, the pancreatic cancer incidence in Jiangsu Province showed an increasing trend. The incidence rate is higher among males and urban residents. It is expected that the incidence of pancreatic cancer will continue to increase in the next decade. Therefore, pancreatic cancer prevention and control efforts should continue to focus on older adults and males. This study develops an advanced provincial prediction model, which provides a quantitative basis for allocating screening resources to high-risk populations and provides a reference paradigm for cancer prevention and control strategies in other developing countries undergoing industrialization.
Collapse
Affiliation(s)
- Hao Yu
- Department of Noncommunicable Chronic Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China
| | - Yuchen Jiang
- Kunshan Center for Disease Control and Prevention, Suzhou, 215100, China
| | - Weigang Miao
- Department of Noncommunicable Chronic Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China
| | - Wenbin Hu
- Kunshan Center for Disease Control and Prevention, Suzhou, 215100, China
| | - Yixu Jin
- Kunshan Center for Disease Control and Prevention, Suzhou, 215100, China
| | - Zhouquan Fan
- Kunshan Center for Disease Control and Prevention, Suzhou, 215100, China
| | - Pengfei Luo
- Department of Noncommunicable Chronic Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China
| | - Ran Tao
- Department of Noncommunicable Chronic Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China
| | - Fangyu Zhu
- Department of Noncommunicable Chronic Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China
| | - Renqiang Han
- Department of Noncommunicable Chronic Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China.
| | - Jinyi Zhou
- Department of Noncommunicable Chronic Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China.
| |
Collapse
|
23
|
Gheybi E, Hosseinzadeh P, Tayebi-Khorrami V, Rostami M, Soukhtanloo M. Proteomics in decoding cancer: A review. Clin Chim Acta 2025; 574:120302. [PMID: 40220985 DOI: 10.1016/j.cca.2025.120302] [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: 03/03/2025] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
Cancer remains the second leading cause of death worldwide, posing a significant global health challenge. Extensive research has revealed common biological characteristics across cancer cells, forming the foundation for developing innovative diagnostic and therapeutic strategies. To better understand these shared traits, advanced measurement technologies are critical. Proteomics, the large-scale study of proteins and their functions, has emerged as a transformative tool for uncovering the complexities of cancer biology. This approach provides an in-depth view of cellular activities and protein interactions, offering unprecedented insights into cancer progression and treatment. Unlike traditional methods that investigate specific pathways in isolation, proteomics enables simultaneous analysis of thousands of proteins, generating a comprehensive understanding of cancer biology. This review explores the mechanisms underlying proteomics, its application to understanding cancer hallmarks, and its potential to transform clinical approaches. By examining proteomics' role in metastasis, angiogenesis, proliferation, and resistance mechanisms, this study highlights its contributions to cancer diagnosis, treatment, and personalized medicine. Additionally, prospects in integrating proteomics with other -omics fields and advancements in computational analysis will be discussed. This work aims to illuminate the path toward more effective, precise, and individualized cancer care through proteomics innovations.
Collapse
Affiliation(s)
- Elaheh Gheybi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pejman Hosseinzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Tayebi-Khorrami
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Rostami
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
24
|
Fan Z, Xiao Y, Du Y, Zhang Y, Zhou W. Pancreatic cancer subtyping - the keystone of precision treatment. Front Immunol 2025; 16:1563725. [PMID: 40264765 PMCID: PMC12011869 DOI: 10.3389/fimmu.2025.1563725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/17/2025] [Indexed: 04/24/2025] Open
Abstract
In recent years, the incidence and mortality rates of pancreatic cancer have been rising, posing a severe threat to human health. Tumor heterogeneity remains a critical barrier to advancing diagnosis and treatment efforts. The lack of specific early symptoms, limited early diagnostic methods, high biological complexity, and restricted therapeutic options contribute to the poor outcomes and prognosis of pancreatic cancer. Therefore, there is an urgent need to explore the different subtypes in-depth and develop personalized therapeutic strategies tailored to each subtype. Increasing evidence highlights the pivotal role of molecular subtyping in treating pancreatic cancer. This review focuses on recent advancements in classifying molecular subtypes and therapeutic approaches, discussed from the perspectives of gene mutations, genomics, transcriptomics, proteomics, metabolomics, and immunomics.
Collapse
Affiliation(s)
- Zeyang Fan
- The Second Clinical Medical School, Lanzhou University,
Lanzhou, China
| | - Yao Xiao
- The Second Clinical Medical School, Lanzhou University,
Lanzhou, China
| | - Yan Du
- The Second Clinical Medical School, Lanzhou University,
Lanzhou, China
| | - Yan Zhang
- The Second Clinical Medical School, Lanzhou University,
Lanzhou, China
| | - Wence Zhou
- Department of General Surgery , The Second Hospital of Lanzhou University & The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| |
Collapse
|
25
|
Balkhi S, Zuccolotto G, Di Spirito A, Rosato A, Mortara L. CAR-NK cell therapy: promise and challenges in solid tumors. Front Immunol 2025; 16:1574742. [PMID: 40260240 PMCID: PMC12009813 DOI: 10.3389/fimmu.2025.1574742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
Over the past few years, cellular immunotherapy has emerged as a promising treatment for certain hematologic cancers, with various CAR-T therapies now widely used in clinical settings. However, challenges related to the production of autologous cell products and the management of CAR-T cell toxicity highlight the need for new cell therapy options that are universal, safe, and effective. Natural killer (NK) cells, which are part of the innate immune system, offer unique advantages, including the potential for off-the-shelf therapy. A recent first-in-human trial of CD19-CAR-NK infusion in patients with relapsed/refractory lymphoid malignancies demonstrated safety and promising clinical activity. Building on these positive clinical outcomes, current research focuses on enhancing CAR-NK cell potency by increasing their in vivo persistence and addressing functional exhaustion. There is also growing interest in applying the successes seen in hematologic malignancies to solid tumors. This review discusses current trends and emerging concepts in the engineering of next-generation CAR- NK therapies. It will cover the process of constructing CAR-NK cells, potential targets for their manufacturing, and their role in various solid tumors. Additionally, it will examine the mechanisms of action and the research status of CAR-NK therapies in the treatment of solid tumors, along with their advantages, limitations, and future challenges. The insights provided may guide future investigations aimed at optimizing CAR-NK therapy for a broader range of malignancies.
Collapse
Affiliation(s)
- Sahar Balkhi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Gaia Zuccolotto
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Anna Di Spirito
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
26
|
Valenzuela-Fuenzalida JJ, Núñez-Castro CI, Morán-Durán VB, Nova-Baeza P, Orellana-Donoso M, Suazo-Santibáñez A, Becerra-Farfan A, Oyanedel-Amaro G, Bruna-Mejias A, Granite G, Casanova-Martinez D, Sanchis-Gimeno J. Anatomical Variants in Pancreatic Irrigation and Their Clinical Considerations for the Pancreatic Approach and Surrounding Structures: A Systematic Review with Meta-Analysis. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:666. [PMID: 40282957 PMCID: PMC12028877 DOI: 10.3390/medicina61040666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: The pancreas receives blood through a complex network of multiple branches, primarily originating from the celiac trunk (CeT) and the superior mesenteric artery (SMA). This blood supply is structured into three main arterial groups, each serving different regions of the pancreas to effectively support its endocrine and exocrine functions. Materials and Methods: The databases Medline, Scopus, Web of Science, Google Scholar, Cumulative Index to Nursing and Allied Health Literature (CINAHL) and Latin American and the Caribbean Literature in Health Sciences (LILACS) were searched until January 2025. Methodological quality was evaluated using an assurance tool for anatomical studies (AQUA). Pooled prevalence was estimated using a random effects model. Results: A total of sixteen studies met the established selection criteria in this study for meta-analysis. Pancreatic irrigation variants presented a prevalence of 11.2% (CI: 7-14%) and a heterogeneity of 88.2%. The other studies were analyzed by subgroups, showing statistically significant differences in the following subgroups: (1) sample type-a larger sample of images analyzed in the included studies (p = 0.312), which did not show statistically significant differences; (2) geographical region (p = 0.041), which showed a greater presence in the Asian population studied, and this was statistically significant; and (3) sex (male or female) (p = 0.12), where there were no statistically significant differences. Conclusions: The discovery of variations in pancreatic irrigation is common due to the numerous blood vessels involved in supplying this vital organ. Understanding different vascular patterns (such as those from the splenic and mesenteric arteries) is crucial for surgical interventions on the pancreas. For transplant patients, a thorough vascular analysis of both the donor and recipient is essential. Variations can impact blood flow and compatibility, potentially leading to transplant rejection if not addressed. To enhance outcomes, it is recommended to develop more accurate imaging tools for pre-surgical analysis, necessitating ongoing research in this area.
Collapse
Affiliation(s)
- Juan José Valenzuela-Fuenzalida
- Departamento de Morfología, Facultad de Medicina, Universidad Andrés Bello, Santiago 8370146, Chile; (J.J.V.-F.); (C.I.N.-C.); (V.B.M.-D.); (P.N.-B.); (A.B.-M.)
| | - Camila Ignacia Núñez-Castro
- Departamento de Morfología, Facultad de Medicina, Universidad Andrés Bello, Santiago 8370146, Chile; (J.J.V.-F.); (C.I.N.-C.); (V.B.M.-D.); (P.N.-B.); (A.B.-M.)
| | - Valeria Belén Morán-Durán
- Departamento de Morfología, Facultad de Medicina, Universidad Andrés Bello, Santiago 8370146, Chile; (J.J.V.-F.); (C.I.N.-C.); (V.B.M.-D.); (P.N.-B.); (A.B.-M.)
| | - Pablo Nova-Baeza
- Departamento de Morfología, Facultad de Medicina, Universidad Andrés Bello, Santiago 8370146, Chile; (J.J.V.-F.); (C.I.N.-C.); (V.B.M.-D.); (P.N.-B.); (A.B.-M.)
| | - Mathias Orellana-Donoso
- Escuela de Medicina, Universidad Finis Terrae, Santiago 7501015, Chile;
- Department of Morphological Sciences, Faculty of Medicine and Science, Universidad San Sebastián, Santiago 7510157, Chile
| | | | - Alvaro Becerra-Farfan
- Departamento de Ciencias Química y Biológicas, Facultad de Ciencias de la Salud, Universidad Bernardo O’Higgins, Santiago 8370993, Chile;
| | - Gustavo Oyanedel-Amaro
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910060, Chile;
| | - Alejandro Bruna-Mejias
- Departamento de Morfología, Facultad de Medicina, Universidad Andrés Bello, Santiago 8370146, Chile; (J.J.V.-F.); (C.I.N.-C.); (V.B.M.-D.); (P.N.-B.); (A.B.-M.)
- Departamento de Ciencias y Geografía, Facultad de Ciencias Naturales y Exactas, Universidad de Playa Ancha, Valparaíso 2360072, Chile
| | - Guinevere Granite
- Department of Surgery, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, ML 20814, USA;
| | - Daniel Casanova-Martinez
- Facultad de Medicina, Universidad de Valparaíso, Campus San Felipe, Valparaíso 2170000, Chile;
- Laboratorio de Neuroanatomía Microquirúrgica (LaNeMic), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1053, Argentina
| | - Juan Sanchis-Gimeno
- GIAVAL Research Group, Department of Anatomy and Human Embryology, Faculty of Medicine, University of Valencia, 46001 Valencia, Spain
| |
Collapse
|
27
|
Haddadin L, Sun X. Stem Cells in Cancer: From Mechanisms to Therapeutic Strategies. Cells 2025; 14:538. [PMID: 40214491 PMCID: PMC11988674 DOI: 10.3390/cells14070538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Stem cells have emerged as a pivotal area of research in the field of oncology, offering new insights into the mechanisms of cancer initiation, progression, and resistance to therapy. This review provides a comprehensive overview of the role of stem cells in cancer, focusing on cancer stem cells (CSCs), their characteristics, and their implications for cancer therapy. We discuss the origin and identification of CSCs, their role in tumorigenesis, metastasis, and drug resistance, and the potential therapeutic strategies targeting CSCs. Additionally, we explore the use of normal stem cells in cancer therapy, focusing on their role in tissue regeneration and their use as delivery vehicles for anticancer agents. Finally, we highlight the challenges and future directions in stem cell research in cancer.
Collapse
Affiliation(s)
| | - Xueqin Sun
- Cancer Genome and Epigenetics Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
28
|
Zhao S, Wang J, Liu H, Liu S, Sun L, Wang Y, Gao S, Sun Y. Analyses of CTNNB1 mutation and expression and clinicopathological characteristics in 179 cases of solid-pseudopapillary neoplasm of the pancreas. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2025; 32:327-338. [PMID: 39991945 DOI: 10.1002/jhbp.12123] [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] [Indexed: 02/25/2025]
Abstract
BACKGROUND Nuclear expression of CTNNB1 is occasionally negative in solid-pseudopapillary neoplasm (SPN) of the pancreas, leading to a missed diagnosis. In the present study, we aimed to investigate the clinical significance of CTNNB1 mutation detection for diagnosing SPN and explore the difference in clinicopathological characteristics at different ages and sex. METHODS Patients who underwent surgery for a pathologically confirmed SPN in our institution between 2011 and 2020 were collected. Their clinicopathological data were analyzed. RESULTS The median age of the 179 patients was 31 years (6-64 years), including 34 pediatric patients (19.0%), and 32 patients were male (17.9%). We detected point mutations in exon 3 of CTNNB1 in 74.3% (133/179) of SPNs by Sanger sequencing. The main mutation sites were D32, S33, S37, G34 and T41. In the three SPNs without nuclear expression of CTNNB1, Sanger sequencing showed point mutations of CTNNB1. NGS did not detect any consistent mutation except CTNNB1 in the three cases. The tumor size, Ki-67 index, and the negative rates of CTNNB1 nuclear expression and synaptophysin expression in the pediatric group were higher than those in other groups (p < .05). CONCLUSIONS For atypical cases, testing for CTNNB1 mutations can help in the accurate diagnosis of SPN. Compared with adult patients, pediatrics with SPN may be more prone to recurrence, and their immunohistochemical phenotype is more complex, requiring additional care in the diagnosis and postoperative follow-up.
Collapse
Affiliation(s)
- Shuai Zhao
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jian Wang
- Department of Pancreatic Carcinoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Huimin Liu
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Shasha Liu
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lin Sun
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ying Wang
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Song Gao
- Department of Pancreatic Carcinoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Yan Sun
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
29
|
Dabiri M, Tehrani M, Rafiei A, Valadan R. Production and functional analysis of a phage displayed scFv recombinant antibody targeting EGFR/HER2 dimerization domain. Protein Expr Purif 2025; 228:106649. [PMID: 39722421 DOI: 10.1016/j.pep.2024.106649] [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: 10/30/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Tumor cells exploit epidermal growth factor receptor (EGFR) family to develop resistance against therapeutic antibodies, such as Herceptin. Upon ligand binding, dimerization between EGFR and HER2 is one of the most important causes of treatment failure in breast cancer and other cancers expressing EGFR and HER2. The aim of this study was to develop and evaluate the function of a human recombinant single-chain variable fragment (scFv) antibody against the dimerization domain of EGFR to inhibit its interaction with other members of the epidermal growth factor receptor family, especially HER2. METHODS scFv against EGFR was expressed and purified. Cell-ELISA, MTT assay, inhibition of STAT3 phosphorylation, quantitative RT-PCR, and dimerization inhibition were performed on EGFR and HER2 expressing cell lines to characterize functional properties of the produced scFv. The conformational structure of the produced scFv and its binding ability to EGFR was computationally investigated. RESULTS In vitro binding analysis by cell-ELISA revealed the EGFR binding ability of the purified antibodies and confirmed by immunoblotting. ScFvs preferentially reduced the proliferation and survival of MCF7, MDA-MB-468, and SKOV3 cell lines with no effect on the VERO line. More considerably, MCF7 cells treated with the scFv antibody showed reduced STAT3 phosphorylation, decreased Bcl-2 expression, and increased Bax expression. Finally, the scFvs hindered EGFR and HER2 dimerization. CONCLUSION The produced scFv antibody showed to be functional in a simultaneous blockade of EGFR and HER2, suggesting its potential as a promising candidate for targeted therapy against various EGFR overexpressing tumors.
Collapse
Affiliation(s)
- Mina Dabiri
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Department of Biomedical and Biological Sciences, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ, 08028, United States.
| | - Mohsen Tehrani
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Alireza Rafiei
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Reza Valadan
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center (MCBRC), School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
30
|
Kim HS, Chae H, Lim SY, Jeong H, Yoon SJ, Shin SH, Han IW, Heo JS, Kim H. Implications of portal vein/superior mesenteric vein involvement in pancreatic cancer: A comprehensive correlation from preoperative radiological assessment to resection, pathology, and long-term outcomes. A retrospective cohort study. Int J Surg 2025; 111:2962-2972. [PMID: 39998570 DOI: 10.1097/js9.0000000000002307] [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: 07/16/2024] [Accepted: 02/03/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND The incidence of portal vein/superior mesenteric vein (PV/SMV) resection during pancreatoduodenectomy is increasing in clinical practice. This study investigated the clinical significance of preoperative PV/SMV assessment and intraoperative resection and their correlation with pathological results and long-term survival outcomes. METHODS We analyzed 443 patients undergoing pancreatoduodenectomy at a tertiary center from 2012 to 2017 based on PV/SMV resection. Subgroup analyses were performed based on preoperative PV/SMV involvement, resection, and margin status. RESULTS Total of 441 patients were analyzed; 175 had PV/SMV involvement on preoperative radiological assessments and 128 underwent PV/SMV resection. True pathological invasion was observed in 78 patients (60.9%), with 34.3% showing no invasion and negative margins. The positive predictive value for preoperative PV/SMV involvement was 61.7%, with a false-negative value of 28.9%. Overall survival of patients who underwent PV/SMV resection was worse than those who did not (2-year survival rate, 38.1% vs 54.9%, P < 0.001). Patients without PV/SMV resection with an rR1/R1 margin showed no decrease in survival compared to those with PV/SMV resection and R0 margins (54.9% vs 40.3%, P = 0.029). Prognostic factors included hypertension, PV/SMV resection, PV/SMV R2 margin, T stage, N stage, cell differentiation, adjuvant treatment, and recurrence. CONCLUSION PV/SMV resection could ensure R0 resection but may lead to unnecessary resection. Careful consideration is essential in determining the need for PV/SMV resection. Poor survival in such patients highlights the need for tailored treatments, including neoadjuvant therapy, for those who are expected to undergo PV/SMV resections.
Collapse
Affiliation(s)
- Hyeong Seok Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hochang Chae
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Soo Yeun Lim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Department of Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - HyeJeong Jeong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Department of Surgery, Daejeon Eulji Medical Center, Eulji University School of Medicine, Daejeon, South Korea
| | - So Jeong Yoon
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sang Hyun Shin
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - In Woong Han
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jin Seok Heo
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hongbeom Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
31
|
Oikonomou D, Bhogal RH, Mavroeidis VK. Central pancreatectomy: An uncommon but potentially optimal choice of pancreatic resection. Hepatobiliary Pancreat Dis Int 2025; 24:119-127. [PMID: 39578167 DOI: 10.1016/j.hbpd.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 11/01/2024] [Indexed: 11/24/2024]
Abstract
Benign, premalignant or low-grade malignant pancreatic tumors are increasingly diagnosed owing to the widespread uptake of cross-sectional imaging. Surgical excision is a potential treatment option for these tumors. Pancreatoduodenectomy and distal pancreatectomy are the standard resections for tumors located in the pancreatic head-neck or body-tail, respectively, and not uncommonly sacrifice a significant amount of healthy pancreatic parenchyma. Central pancreatectomy (CP) is a parenchyma-sparing procedure, initially performed by Dagradi and Serio in 1982, in a patient with pancreatic neck insulinoma. Since then, an increasing number of cases are being performed worldwide, either via open or minimally invasive surgical access. Additionally, pancreatic enucleation is reserved for tumors < 3 cm, without involvement of the main pancreatic duct. CP remains an alternative approach in selected cases, albeit in the presence of some controversies, such as its use in early pancreatic ductal adenocarcinoma or metastatic deposits to the central aspect of the pancreas from other malignancies. In recent years, clarity is lacking as regards indications for CP, and despite accumulating evidence in favor of limited resections for suitable pancreatic tumors, no evidence-based consensus guidelines are yet available. Nevertheless, it appears that appropriate patient selection is of paramount importance to maximize the advantages of preservation of endocrine and exocrine pancreatic functions as well as to mitigate the risks of higher complication rates. In this comprehensive review, we explore the role of CP in the treatment of lesions located in the neck and proximal body of the pancreas.
Collapse
Affiliation(s)
- Dimitrios Oikonomou
- Department of HPB Surgery, King's College Hospital, Denmark Hill, London, SE5 9RS, UK
| | - Ricky H Bhogal
- Department of Academic Surgery, The Royal Marsden NHS Foundation Trust, Fulham Road, Chelsea, London SW3 6JJ, UK
| | - Vasileios K Mavroeidis
- Department of Academic Surgery, The Royal Marsden NHS Foundation Trust, Fulham Road, Chelsea, London SW3 6JJ, UK; Department of HPB Surgery, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol Royal Infirmary, Upper Maudlin St, Bristol BS2 8HW, UK.
| |
Collapse
|
32
|
Wu Y, Zhang C, Huang J, Chen Q, Zhang Y, Liu F, Xu D, Jiang K, Shi R, Chen M, Yuan H. Integrated analysis of scRNA-seq and bulk RNA-seq data identifies BHLHE40 as a key gene in pancreatic cancer progression and gemcitabine resistance. Semin Oncol 2025; 52:152338. [PMID: 40250076 DOI: 10.1016/j.seminoncol.2025.152338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 04/20/2025]
Abstract
OBJECTIVE Pancreatic cancer is characterized by its high mortality rate and short survival periods, and novel therapeutic targets and tailor personalized strategies are urgently needed. In this study, we aim to investigate the molecular mechanisms underlying pancreatic ductal adenocarcinoma (PDAC) progression and chemoresistance, with a focus on identifying novel therapeutic targets. METHODS Multiomics approaches were integrated to identify novel actionable targets for PDAC. Public datasets such as TCGA and GEO were utilized to investigate the relationship between gene expression and clinical outcomes. Functional enrichment, cell-cell communication, and metabolic pathway analyses were performed to reveal PDAC heterogeneity and therapeutic resistance mechanisms. RESULTS BHLHE40 was identified as a hub gene linked to high-CNV PDAC cells, Gemcitabine resistance, and poor prognosis in PDAC. High BHLHE40 expression is significantly correlated with immunosuppressive tumor microenvironment (TME) features such as reduced CD8+ T infiltration, TCR richness, and lower tumor mutational burden (TMB). ChIP-seq data analysis confirmed BHLHE40 could directly bind to the SAT1 promoter, establishing a transcriptional axis promoting chemoresistance. Single-cell RNA-seq analysis further revealed that the BHLHE40+/SAT1+ subpopulation cells are resistant to Gemcitabine in PDAC. CONCLUSIONS BHLHE40 is significantly correlated with PDAC malignancy and chemoresistance via SAT1 regulation and immune evasion. Targeting BHLHE40 may sensitize PDACs to Gemcitabine and facilitate personalized treatment for BHLHE40+ PDAC patients.
Collapse
Affiliation(s)
- Yang Wu
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chun Zhang
- Department of Gastroenterology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiacheng Huang
- Department of Urology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qun Chen
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yufeng Zhang
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fengyuan Liu
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dong Xu
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kuirong Jiang
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mengxing Chen
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Yuan
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
33
|
Wannberg F, Hjalmar V, Ng H, Johansson C, Probert F, Phillipson M, Åberg M, Gordon M, Mackman N, Rosell A, Thålin C. Plasma H3Cit-DNA Discriminates Between Cancer and Inflammation in a Cohort of Patients with Unspecific Cancer Symptoms. Inflammation 2025; 48:760-769. [PMID: 38941006 PMCID: PMC12053196 DOI: 10.1007/s10753-024-02085-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
Cancer detection is challenging, especially in patients with unspecific cancer symptoms. Biomarkers could identify patients at high risk of cancer. Prior studies indicate that neutrophil extracellular traps (NETs) are associated with cancer, but also with autoimmune and infectious diseases. The objective of this prospective study was to investigate markers associated with NET formation (nucleosomal citrullinated histone 3 [H3Cit-DNA], cell free DNA [cfDNA] and neutrophil elastase [NE]), and c-reactive protein (CRP) in patients with unspecific cancer symptoms, such as fatigue, weight loss or radiological sign of malignancy without an apparent primary tumor, referred to the Diagnostic Center at Danderyd Hospital in Sweden. Blood samples were drawn on admission, before cancer diagnosis. Out of 475 patients, 160 (34%) were diagnosed with cancer, 56 (12%) with autoimmune disease, 32 (7%) with infectious disease, 71 (15%) with other diseases and 156 (33%) received no diagnosis. H3Cit-DNA, cfDNA, NE and CRP were significantly higher in patients with cancer compared to patients without cancer (p < 0.0001, p < 0.0001, p = 0.004, and p = 0.0002 respectively). H3Cit-DNA, but not cfDNA, NE or CRP, was significantly elevated in patients with cancer compared to patients with autoimmune disease (p = 0.0001). H3Cit-DNA, cfDNA, NE or CRP did not differ between cancer and infectious disease. In conclusion, H3Cit-DNA is elevated in patients diagnosed with cancer compared to non-cancer patients with the same symptomatology. Further studies should evaluate if H3Cit-DNA could aid in selecting patients that would benefit the most from a rapid cancer diagnostic work-up.
Collapse
Affiliation(s)
- Fredrika Wannberg
- Department of Clinical Sciences, Division of Internal Medicine, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Viktoria Hjalmar
- Department of Clinical Sciences, Division of Internal Medicine, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- Division of Specialist Medical Care, Danderyd Hospital, Diagnostic Center, Stockholm, Sweden
| | - Henry Ng
- Department of Clinical Sciences, Division of Internal Medicine, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Cell Biology, SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Caroline Johansson
- Division of Specialist Medical Care, Danderyd Hospital, Diagnostic Center, Stockholm, Sweden
| | - Fay Probert
- Department of Chemistry, University of Oxford, Oxford, UK
| | - Mia Phillipson
- Department of Medical Cell Biology, SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry and SciLifeLab Affinity Proteomics, Uppsala University, Uppsala, Sweden
| | - Max Gordon
- Division of Internal Medicine, Department of Clinical Sciences, Division of Orthopedics, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Nigel Mackman
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Axel Rosell
- Department of Clinical Sciences, Division of Internal Medicine, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Charlotte Thålin
- Department of Clinical Sciences, Division of Internal Medicine, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
34
|
Cai W, Zhu Y, Li D, Hu M, Teng Z, Cong R, Chen Z, Sun X, Ma X, Zhao X. Baseline Body Composition and 3D-Extracellular Volume Fraction for Predicting Pancreatic Fistula after Distal Pancreatectomy in Pancreatic Body and/or Tail Adenocarcinoma. Acad Radiol 2025; 32:2027-2040. [PMID: 39537519 DOI: 10.1016/j.acra.2024.10.010] [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: 05/22/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024]
Abstract
RATIONALE AND OBJECTIVES Clinically relevant postoperative pancreatic fistula (CR-POPF) is a threatening complication in body and/or tail pancreatic ductal adenocarcinoma (PDAC) receiving distal pancreatectomy (DP) and is difficult to predict preoperatively. We aimed to identify the role of baseline CT-based body composition analysis and extracellular volume (ECV) map in predicting the risk of CR-POPF preoperatively. MATERIALS AND METHODS A total of 329 resectable PDAC patients were enrolled and underwent multiphasic contrast-enhanced CT. Body composition indicators were calculated, and ECV maps were generated through multiphasic CT images. The differences in clinical variables and quantitative parameters between CR-POPF and non-CR-POPF patients were compared. Correlations between ECV fraction and pancreatic fibrosis stage were analyzed. Multivariate logistic regression was performed to screen the independent predictors and develop prediction models for CR-POPF. Receiver operating characteristic curve was utilized to evaluate the predictive performance. RESULTS Among 329 patients, 19.76% (65/329) developed CR-POPF. Albumin, pancreatic texture, and intraoperative blood loss were used to build the clinical model with an AUC of 0.764. ECV fraction and total muscle ratio (TMR) were chosen to build the radiological model with an AUC of 0.872. A combined nomogram integrated with albumin, ECV fraction, and TMR could significantly improve the discrimination ability to an AUC of 0.924 (Delong test, all p < 0.05). The ECV fraction showed high positive correlation with histological fibrosis grade (Spearman ρ = 0.81). CONCLUSION CT-based body composition analysis and ECV exhibited great potential for predicting CR-POPF in body and/or tail PDAC after DP. The combined nomogram could further improve the predictive performance.
Collapse
Affiliation(s)
- Wei Cai
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Yongjian Zhu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Dengfeng Li
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Mancang Hu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Ze Teng
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Rong Cong
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Zhaowei Chen
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Xujie Sun
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (X.S.).
| | - Xiaohong Ma
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| | - Xinming Zhao
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (W.C., Y.Z., D.L., M.H., Z.T., R.C., Z.C., X.M., X.Z.).
| |
Collapse
|
35
|
Lu XY, Tan XD. Clinical outcomes of interlocking main pancreatic duct-jejunal internal bridge drainage in middle pancreatectomy: A comparative study. World J Gastrointest Surg 2025; 17:102428. [PMID: 40162424 PMCID: PMC11948093 DOI: 10.4240/wjgs.v17.i3.102428] [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: 10/31/2024] [Revised: 12/01/2024] [Accepted: 01/16/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Middle pancreatectomy (MP) is a surgical procedure that removes non-invasive lesions in the pancreatic neck and body, allowing for the preservation of pancreatic function. However, MP is associated with a higher risk of postoperative complications, and there's no clear consensus on which anastomotic method is preferable. In recent years, our team has developed a new method called interlocking main pancreatic duct-jejunal (IMPD-J) internal bridge drainage to MP. AIM To compare perioperative and postoperative outcomes in patients who underwent IMPD-J bridge drainage and those underwent traditional duct-to-mucosa pancreatojejunostomy. METHODS Patients who underwent MP in our hospital between October 1, 2011 and July 31, 2023 were enrolled in this study. Patients were divided into two groups based on their pancreatojejunostomy technique: IMPD-J bridge drainage group and duct-to-mucosa pancreatojejunostomy group. Demographic data (age, gender, body mass index, hypertension, diabetes, etc.) and perioperative indicators [operation time, intraoperative bleeding, clinically relevant postoperative pancreatic fistula (CR-POPF), delayed gastric emptying, etc.] were recorded and analyzed statistically. RESULTS A total of 53 patients were enrolled in this study, including 23 in the IMPD-J Bridge Drainage group and 30 in the traditional duct-to-mucosa pancreatojejunostomy group. There were no significant differences in demographic or preoperative characteristics between the groups. Compared to traditional duct-to-mucosa pancreaticojejunostomy, IMPD-J bridge drainage had a significant shorter operation time (4.3 ± 1.3 hours vs 5.8 ± 1.8 hours, P = 0.002), nasogastric tube retention days (5.3 ± 1.7 days vs 6.5 ± 2.0 days, P = 0.031), lower incidence of delayed gastric emptying (8.7% vs 36.7%, P = 0.019), and lower incidence of CR-POPF (39.1% vs 70.0%, P = 0.025). Multivariate logistic regression analysis showed that pancreaticojejunostomy type (odds ratio = 4.219, 95% confidence interval = 1.238-14.379, P = 0.021) and plasma prealbumin (odds ratio = 1.132, 95% confidence interval = 1.001-1.281, P = 0.049) were independent risk factor for CR-POPF. In IMPD-J bridge drainage group, only one patient experienced recurrent pancreatitis due to the large diameter of the silicone tube and had it removed six months after surgery. CONCLUSION Compared to traditional duct-to-mucosa pancreatojejunostomy, IMPD-J bridge drainage has the advantages of simplicity and fewer perioperative complications, with favorable long-term outcomes.
Collapse
Affiliation(s)
- Xin-Yan Lu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Xiao-Dong Tan
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| |
Collapse
|
36
|
Farinella R, Felici A, Peduzzi G, Testoni SGG, Costello E, Aretini P, Blazquez-Encinas R, Oz E, Pastore A, Tacelli M, Otlu B, Campa D, Gentiluomo M. From classical approaches to artificial intelligence, old and new tools for PDAC risk stratification and prediction. Semin Cancer Biol 2025; 112:71-92. [PMID: 40147701 DOI: 10.1016/j.semcancer.2025.03.004] [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: 11/28/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is recognized as one of the most lethal malignancies, characterized by late-stage diagnosis and limited therapeutic options. Risk stratification has traditionally been performed using epidemiological studies and genetic analyses, through which key risk factors, including smoking, diabetes, chronic pancreatitis, and inherited predispositions, have been identified. However, the multifactorial nature of PDAC has often been insufficiently addressed by these methods, leading to limited precision in individualized risk assessments. Advances in artificial intelligence (AI) have been proposed as a transformative approach, allowing the integration of diverse datasets-spanning genetic, clinical, lifestyle, and imaging data into dynamic models capable of uncovering novel interactions and risk profiles. In this review, the evolution of PDAC risk stratification is explored, with classical epidemiological frameworks compared to AI-driven methodologies. Genetic insights, including genome-wide association studies and polygenic risk scores, are discussed, alongside AI models such as machine learning, radiomics, and deep learning. Strengths and limitations of these approaches are evaluated, with challenges in clinical translation, such as data scarcity, model interpretability, and external validation, addressed. Finally, future directions are proposed for combining classical and AI-driven methodologies to develop scalable, personalized predictive tools for PDAC, with the goal of improving early detection and patient outcomes.
Collapse
Affiliation(s)
| | | | | | - Sabrina Gloria Giulia Testoni
- Division of Gastroenterology and Gastrointestinal Endoscopy, IRCCS Policlinico San Donato, Vita-Salute San Raffaele University, Milan, Italy
| | - Eithne Costello
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Paolo Aretini
- Fondazione Pisana per la Scienza, San Giuliano Terme, Italy
| | - Ricardo Blazquez-Encinas
- Department of Cell Biology, Physiology and Immunology, University of Cordoba / Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Elif Oz
- Department of Biostatistics and Bioinformatics, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Aldo Pastore
- Fondazione Pisana per la Scienza, San Giuliano Terme, Italy
| | - Matteo Tacelli
- Pancreas Translational & Clinical Research Center, Pancreato-Biliary Endoscopy and Endosonography Division, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Burçak Otlu
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, Ankara, Turkey
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | | |
Collapse
|
37
|
Liu B, Zhang X, Li J, Sun Z, Lin C, Ning C, Hong X, Zhu S, Shen D, Chen L, Huang G. The Global, Regional, and National Burden of Pancreatitis in 204 Countries and Territories, 1990-2021: A Systematic Analysis for the Global Burden of Disease Study 2021. Dig Dis Sci 2025:10.1007/s10620-025-08996-y. [PMID: 40140166 DOI: 10.1007/s10620-025-08996-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/14/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Pancreatitis, an inflammatory condition of the pancreas, poses a significant global health burden. This study provides up-to-date global, regional, and national estimates of pancreatitis burdens from 1990 to 2021, focusing on disparities and trends across regions, age groups, and sexes. METHODS Data from the Global Burden of Disease (GBD) 2021 study were analyzed to assess the incidence, prevalence, mortality, and disability-adjusted life years (DALYs) associated with pancreatitis. Trends over the 32-year period were examined across demographics and geographic regions. Average annual percentage changes (AAPC) with 95% confidence intervals (CI) were calculated. RESULTS In 2021, the age-standardized rates (95% uncertainty interval) per 100,000 population for pancreatitis were: prevalence 69.0 (51.3, 91.3), incidence 32.8 (28.9, 37.4), deaths 1.5 (1.3, 1.7), and DALYs 48.4 (43.1, 55.3). Eastern Europe had the highest burden, led by the Russian Federation. From 1990 to 2021, global pancreatitis burden decreased with AAPCs (95% CI) of - 1.0% (- 1.1%, - 1.0%) for prevalence, - 0.4% (- 0.5%, - 0.4%) for incidence, - 0.5% (- 0.6%, - 0.3%) for deaths, and - 0.5% (- 0.6%, - 0.4%) for DALYs. Most regions showed declines, except Eastern Europe and parts of Africa. The elderly, particularly those aged 65 and older, faced the highest burden. An upward incidence trend was noted in those aged 15 to 25. Men exhibited higher burden rates, with the peak burden occurring at younger ages compared to women. High alcohol use contributed to 15.2% of deaths and 17.0% of DALYs related to pancreatitis. CONCLUSIONS This study offers a comprehensive assessment of the global burden of pancreatitis from 1990 to 2021. Despite an overall decline, significant regional and demographic disparities persist, with Eastern Europe remaining disproportionately affected. The high burden among the elderly and rising incidence among the young highlight the need for targeted prevention, early detection, and management strategies.
Collapse
Affiliation(s)
- Baiqi Liu
- Division of Pancreatic Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- Division of Hernia and Abdominal Wall Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- FuRong Laboratory, Changsha, 410078, Hunan Province, China
| | - Xinge Zhang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiarong Li
- Division of Pancreatic Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- Division of Hernia and Abdominal Wall Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- FuRong Laboratory, Changsha, 410078, Hunan Province, China
| | - Zefang Sun
- Division of Pancreatic Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- Division of Hernia and Abdominal Wall Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- FuRong Laboratory, Changsha, 410078, Hunan Province, China
| | - Chiayen Lin
- Division of Pancreatic Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- Division of Hernia and Abdominal Wall Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- FuRong Laboratory, Changsha, 410078, Hunan Province, China
| | - Caihong Ning
- Division of Pancreatic Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- Division of Hernia and Abdominal Wall Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- FuRong Laboratory, Changsha, 410078, Hunan Province, China
| | - Xiaoyue Hong
- Division of Pancreatic Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- Division of Hernia and Abdominal Wall Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- FuRong Laboratory, Changsha, 410078, Hunan Province, China
| | - Shuai Zhu
- Division of Pancreatic Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- Division of Hernia and Abdominal Wall Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- FuRong Laboratory, Changsha, 410078, Hunan Province, China
| | - Dingcheng Shen
- Division of Pancreatic Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- Division of Hernia and Abdominal Wall Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- FuRong Laboratory, Changsha, 410078, Hunan Province, China
| | - Lu Chen
- Division of Pancreatic Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- Division of Hernia and Abdominal Wall Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- FuRong Laboratory, Changsha, 410078, Hunan Province, China
| | - Gengwen Huang
- Division of Pancreatic Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China.
- Division of Hernia and Abdominal Wall Surgery, Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China.
- FuRong Laboratory, Changsha, 410078, Hunan Province, China.
| |
Collapse
|
38
|
Daniel N, Farinella R, Belluomini F, Fajkic A, Rizzato C, Souček P, Campa D, Hughes DJ. The relationship of the microbiome, associated metabolites and the gut barrier with pancreatic cancer. Semin Cancer Biol 2025; 112:43-57. [PMID: 40154652 DOI: 10.1016/j.semcancer.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Pancreatic cancers have high mortality and rising incidence rates which may be related to unhealthy western-type dietary and lifestyle patterns as well as increasing body weights and obesity rates. Recent data also suggest a role for the gut microbiome in the development of pancreatic cancer. Here, we review the experimental and observational evidence for the roles of the oral, gut and intratumoural microbiomes, impaired gut barrier function and exposure to inflammatory compounds as well as metabolic dysfunction as contributors to pancreatic disease with a focus on pancreatic ductal adenocarcinoma (PDAC) initiation and progression. We also highlight some emerging gut microbiome editing techniques currently being investigated in the context of pancreatic disease. Notably, while the gut microbiome is significantly altered in PDAC and its precursor diseases, its utility as a diagnostic and prognostic tool is hindered by a lack of reproducibility and the potential for reverse causality in case-control cohorts. Future research should emphasise longitudinal and mechanistic studies as well as integrating lifestyle exposure and multi-omics data to unravel complex host-microbiome interactions. This will allow for deeper aetiologic and mechanistic insights that can inform treatments and guide public health recommendations.
Collapse
Affiliation(s)
- Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | | | | | - Almir Fajkic
- Department of Pathophysiology Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | | | - Pavel Souček
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic; Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland.
| |
Collapse
|
39
|
Wang J, Wang H, Zou F, Gu J, Deng S, Cao Y, Cai K. The Role of Inorganic Nanomaterials in Overcoming Challenges in Colorectal Cancer Diagnosis and Therapy. Pharmaceutics 2025; 17:409. [PMID: 40284405 PMCID: PMC12030334 DOI: 10.3390/pharmaceutics17040409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/12/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Colorectal cancer poses a significant threat to human health due to its high aggressiveness and poor prognosis. Key factors impacting patient outcomes include post-surgical recurrence, chemotherapeutic drug resistance, and insensitivity to immunotherapy. Consequently, early diagnosis and the development of effective targeted therapies are essential for improving prevention and treatment strategies. Inorganic nanomaterials have gained prominence in the diagnosis and treatment of colorectal cancer owing to their unique size, advantageous properties, and high modifiability. Various types of inorganic nanomaterials-such as metal-based, metal oxide, quantum dots, magnetic nanoparticles, carbon-based, and rare-earth nanomaterials-have demonstrated significant potential in enhancing multimodal imaging, drug delivery, and synergistic therapies. These advancements underscore their critical role in improving therapeutic outcomes. This review highlights the properties and development of inorganic nanomaterials, summarizes their recent applications and progress in colorectal cancer diagnosis and treatment, and discusses the challenges in translating these materials into clinical use. It aims to provide valuable insights for future research and the clinical application of inorganic nanomaterials in colorectal cancer management.
Collapse
Affiliation(s)
- Jun Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (J.W.); (H.W.); (F.Z.)
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Hanwenchen Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (J.W.); (H.W.); (F.Z.)
| | - Falong Zou
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (J.W.); (H.W.); (F.Z.)
| | - Junnan Gu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Shenghe Deng
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Yinghao Cao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Kailin Cai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (J.W.); (H.W.); (F.Z.)
| |
Collapse
|
40
|
Chatterjee A, Prado R, Siddiki H, Stevens T. Clinical Evaluation of Patients with Elevated Serum Lipase. Dig Dis Sci 2025:10.1007/s10620-025-08929-9. [PMID: 40119240 DOI: 10.1007/s10620-025-08929-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 02/14/2025] [Indexed: 03/24/2025]
Affiliation(s)
- Arjun Chatterjee
- Department of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Renan Prado
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Hassan Siddiki
- Department of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Tyler Stevens
- Department of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
| |
Collapse
|
41
|
Ho M, Paruzzo L, Minehart J, Nabar N, Noll JH, Luo T, Garfall A, Zanwar S. Extramedullary Multiple Myeloma: Challenges and Opportunities. Curr Oncol 2025; 32:182. [PMID: 40136386 PMCID: PMC11940950 DOI: 10.3390/curroncol32030182] [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: 02/11/2025] [Revised: 03/09/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025] Open
Abstract
Extramedullary multiple myeloma (EMM), defined in this review as soft tissue plasmacytomas resulting from hematogenous spread, is characterized by the ability of MM cells to proliferate outside of the bone marrow microenvironment. It is aggressive, often associated with high-risk cytogenetics and early relapse, and independently portends significantly shorter progression-free and overall survival, even in the era of highly effective immunotherapies. The molecular and microenvironmental factors underlying extramedullary MM dissemination continue to be studied to inform the development of better treatments. In this review, we discuss our current understanding of the biology of EMM, focusing on its distinct molecular and microenvironmental characteristics vis-à-vis MM. We also review the current treatment strategies, acknowledging the paucity of large, randomized studies specific to this population.
Collapse
Affiliation(s)
- Matthew Ho
- Division of Hematology, University of Pennsylvania, Philadelphia, PA 19104, USA; (M.H.); (L.P.); (J.M.); (N.N.); (J.H.N.); (T.L.); (A.G.)
| | - Luca Paruzzo
- Division of Hematology, University of Pennsylvania, Philadelphia, PA 19104, USA; (M.H.); (L.P.); (J.M.); (N.N.); (J.H.N.); (T.L.); (A.G.)
| | - Janna Minehart
- Division of Hematology, University of Pennsylvania, Philadelphia, PA 19104, USA; (M.H.); (L.P.); (J.M.); (N.N.); (J.H.N.); (T.L.); (A.G.)
| | - Neel Nabar
- Division of Hematology, University of Pennsylvania, Philadelphia, PA 19104, USA; (M.H.); (L.P.); (J.M.); (N.N.); (J.H.N.); (T.L.); (A.G.)
| | - Julia Han Noll
- Division of Hematology, University of Pennsylvania, Philadelphia, PA 19104, USA; (M.H.); (L.P.); (J.M.); (N.N.); (J.H.N.); (T.L.); (A.G.)
| | - Thomas Luo
- Division of Hematology, University of Pennsylvania, Philadelphia, PA 19104, USA; (M.H.); (L.P.); (J.M.); (N.N.); (J.H.N.); (T.L.); (A.G.)
| | - Alfred Garfall
- Division of Hematology, University of Pennsylvania, Philadelphia, PA 19104, USA; (M.H.); (L.P.); (J.M.); (N.N.); (J.H.N.); (T.L.); (A.G.)
| | - Saurabh Zanwar
- Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
42
|
Kataki AD, Gupta PG, Cheema U, Nisbet A, Wang Y, Kocher HM, Pérez-Mancera PA, Velliou EG. Mapping Tumor-Stroma-ECM Interactions in Spatially Advanced 3D Models of Pancreatic Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:16708-16724. [PMID: 40052705 PMCID: PMC11931495 DOI: 10.1021/acsami.5c02296] [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: 02/03/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/21/2025]
Abstract
Bioengineering-based in vitro tumor models are increasingly important as tools for studying disease progression and therapy response for many cancers, including the deadly pancreatic ductal adenocarcinoma (PDAC) that exhibits a tumor/tissue microenvironment of high cellular/biochemical complexity. Therefore, it is crucial for in vitro models to capture that complexity and to enable investigation of the interplay between cancer cells and factors such as extracellular matrix (ECM) proteins or stroma cells. Using polyurethane (PU) scaffolds, we performed a systematic study on how different ECM protein scaffold coatings impact the long-term cell evolution in scaffolds containing only cancer or only stroma cells (activated stellate and endothelial cells). To investigate potential further changes in those biomarkers due to cancer-stroma interactions, we mapped their expression in dual/zonal scaffolds consisting of a cancer core and a stroma periphery, spatially mimicking the fibrotic/desmoplastic reaction in PDAC. In our single scaffolds, we observed that the protein coating affected the cancer cell spatial aggregation, matrix deposition, and biomarker upregulation in a cell-line-dependent manner. In single stroma scaffolds, different levels of fibrosis/desmoplasia in terms of ECM composition/quantity were generated depending on the ECM coating. When studying the evolution of cancer and stroma cells in our dual/zonal model, biomarkers linked to cell aggressiveness/invasiveness were further upregulated by both cancer and stroma cells as compared to single scaffold models. Collectively, our study advances the understanding of how different ECM proteins impact the long-term cell evolution in PU scaffolds. Our findings show that within our bioengineered models, we can stimulate the cells of the PDAC microenvironment to develop different levels of aggressiveness/invasiveness, as well as different levels of fibrosis. Furthermore, we highlight the importance of considering spatial complexity to map cell invasion. Our work contributes to the design of in vitro models with variable, yet biomimetic, tissue-like properties for studying the tumor microenvironment's role in cancer progression.
Collapse
Affiliation(s)
- Anna-Dimitra Kataki
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
| | - Priyanka G. Gupta
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
- School
of Life and Health Sciences, Whitelands College, University of Roehampton, London SW15 4JD, U.K.
| | - Umber Cheema
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
| | - Andrew Nisbet
- Department
of Medical Physics and Biomedical Engineering, University College London, London WC1E 6BT, U.K.
| | - Yaohe Wang
- Centre
for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, U.K.
| | - Hemant M. Kocher
- Centre
for Tumour Biology and Experimental Cancer Medicine, Barts Cancer
Institute, Queen Mary University of London, London EC1M 6BQ, U.K.
| | - Pedro A. Pérez-Mancera
- Department
of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3GE, U.K.
| | - Eirini G. Velliou
- Centre
for 3D models of Health and Disease, Division of Surgery and Interventional
Science, University College London, London W1W 7TY, U.K.
| |
Collapse
|
43
|
Willink CY, Jenniskens SFM, Klaassen NJM, Stommel MWJ, van Laarhoven CJHM, Fütterer JJ, Nijsen JFW. Development of an Intratumoral Holmium Microsphere Injection Method in Ex Vivo Human Pancreatic Ductal Adenocarcinoma: A Preclinical Feasibility Study. Cancers (Basel) 2025; 17:1028. [PMID: 40149361 PMCID: PMC11941289 DOI: 10.3390/cancers17061028] [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: 01/10/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Patients diagnosed with pancreatic ductal adenocarcinoma (PDAC) have a poor prognosis. Local therapy may enhance tumor control and increase resectability. Intratumoral injection of radioactive holmium-166 microspheres presents a promising and minimally invasive treatment with multimodality imaging capabilities (SPECT, CT, MRI). However, holmium-166 microspheres are not commonly used for intratumoral injections, and PDAC is notorious for its high intratumoral pressure. This study developed an intratumoral injection method with nonradioactive holmium-165 microspheres in ex vivo human PDAC specimens using a novel injection system for suspension homogenization. METHODS An injection system was developed and validated in a laboratory setting. Thereafter, intratumoral injections in surgically removed ex vivo PDACs were performed, and parameters were established to optimize feasibility, defined by the ability to inject and control the microsphere distribution. Also, injection limitations and cutoff values were determined. The distribution was assessed by visual confirmation, CT, MRI, ultrasound, and histopathology. RESULTS With a validated injection system, intratumoral injections were performed in ten ex vivo PDAC samples. Feasible injection guidelines include but are not limited to ultrasound or CT needle guidance, a maximum injection volume of <20.0% from the tumor volume, ≤3 needle positions, and an injection volume of 0.3-1.0 mL per needle position. CONCLUSIONS Intratumoral injection of holmium-165 microspheres in ex vivo pancreatic ductal adenocarcinoma was feasible with adherence to injection parameters necessary for effective intratumoral deposition and minimal leakage. The injection system and parameters developed here provide a foundation for future studies on holmium-166 microsphere injections in pancreatic cancer patients, with the aim to improve local tumor control as a part of a multimodal therapy.
Collapse
Affiliation(s)
- Coen Ysbrand Willink
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein-Zuid 10, Postbox 9101, 6500 HB Nijmegen, The Netherlands (J.F.W.N.)
| | - Sjoerd Franciscus Maria Jenniskens
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein-Zuid 10, Postbox 9101, 6500 HB Nijmegen, The Netherlands (J.F.W.N.)
| | - Nienke Johanna Maria Klaassen
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein-Zuid 10, Postbox 9101, 6500 HB Nijmegen, The Netherlands (J.F.W.N.)
| | - Martijn Willem Jan Stommel
- Department of Surgery, Radboud University Medical Center, Geert Grooteplein-Zuid 10, Postbox 9101, 6500 HB Nijmegen, The Netherlands
| | | | - Jurgen J. Fütterer
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein-Zuid 10, Postbox 9101, 6500 HB Nijmegen, The Netherlands (J.F.W.N.)
| | - Johannes Frank Wilhelmus Nijsen
- Department of Medical Imaging, Radboud University Medical Center, Geert Grooteplein-Zuid 10, Postbox 9101, 6500 HB Nijmegen, The Netherlands (J.F.W.N.)
| |
Collapse
|
44
|
Chu MQ, Zhang TJ, Liu ZQ, Yang Q, Du TT, Zhang MJ, Jin Y, Cao YJ, Wen XM, Xu ZJ, Zhao YJ, Lin J, Qian J, Zhou JD. MiR-218 Exhibits Anti-Leukemia Effects by Targeting CTNND2 in Primary Acute Erythroid Leukemia HEL Cells. Cell Biochem Biophys 2025:10.1007/s12013-025-01722-5. [PMID: 40100342 DOI: 10.1007/s12013-025-01722-5] [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] [Accepted: 03/07/2025] [Indexed: 03/20/2025]
Abstract
Acute erythroid leukemia (AEL) is a rare acute myeloid leukemia (AML) subtype that is highly aggressive and is associated with a poor prognosis. Notably, the blockage of erythroid differentiation represents a significant factor in the pathogenesis of erythroleukemia. Prior studies indicated that miR-218 inhibited the erythroid differentiation in a chronic myeloid leukemia (CML)-derived erythroleukemia cell line K562. However, functions of miR-218 in primary AEL remains to be elucidated. To address this gap, functions of miR-218 in HEL cells were evaluated through cell differentiation, cell proliferation, colony formation, cell cycle and cell apoptosis experiments. Subsequently, the targeted downstream genes of miR-218 were identified by the transcriptome sequencing and bioinformatic research, of which demonstrated by the dual-luciferase reporter experiment. Finally, the underlying mechanism of miR-218 in leukemogenesis was identified by enrichment analysis and was validated by western blot (WB) assays. Intriguingly, enhanced miR-218 showed no effect on the erythroid differentiation in HEL cells by determination of the expression of erythroid markers including GATA1, KLF1, TFRC and GYPA. However, miR-218 overexpression in HEL cells presented a markedly anti-proliferative and pro-apoptotic effects, inhibited colony formation and G0/G1 arrest. Transcriptome sequencing and bioinformatics analysis revealed that CTNND2 as the candidate gene of miR-218 within its 3'-untranslated region (3'-UTR) could be bonded by it. Reduced expression level of CTNND2 was further demonstrated by quantitative-PCR and WB after miR-218 overexpression in HEL cells. Furthermore, the luciferase report assay revealed that the CTNND2 production was reduced with its 3'-UTR region was bonded by miR-218. In addition, MAPK signaling pathway was identified and validated as the potential functional pathway involved in leukemogenesis caused by miR-218 overexpression in HEL cells. In summary, miR-218 exhibits anti-proliferative and pro-apoptotic functions by targeting CTNND2 and modulating MAPK signaling in HEL cells, yet it has no impact on the erythroid differentiation process.
Collapse
Affiliation(s)
- Ming-Qiang Chu
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Ting-Juan Zhang
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Department of Oncology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Zi-Qi Liu
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
- Department of Nephrology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Qian Yang
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Ting-Ting Du
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Min-Jie Zhang
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Ye Jin
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Yong-Jie Cao
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Xiang-Mei Wen
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Zi-Jun Xu
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Yang-Jing Zhao
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Jiang Lin
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
- Laboratory Center, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Jun Qian
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China
| | - Jing-Dong Zhou
- Department of Hematology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China.
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China.
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, People's Republic of China.
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Zhenjiang, Jiangsu, People's Republic of China.
| |
Collapse
|
45
|
Sapkota A, Paudel R, Pandey S, Bhatt N. Solid pseudopapillary neoplasm of the pancreas in an adolescent: A case report and review of the literature. World J Gastrointest Oncol 2025; 17:101859. [PMID: 40092930 PMCID: PMC11866218 DOI: 10.4251/wjgo.v17.i3.101859] [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: 09/28/2024] [Revised: 11/27/2024] [Accepted: 12/19/2024] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Solid pseudopapillary neoplasm (SPN) of the pancreas is a rare epithelial tumor that primarily affects young women. Since the condition is often asymptomatic or presents with non-specific symptoms, its diagnosis can be difficult. CASE SUMMARY This report details the case of a 15-year-old girl who presented with a 2-year history of abdominal pain, with no significant findings during physical examination. Abdominal ultrasound revealed a well-defined heterogeneous solid-cystic mass in the epigastric region, likely originating from the tail of the pancreas. A subsequent contrast-enhanced computed tomography scan indicated a well-defined cystic lesion with an enhancing solid component and capsule in the tail of the pancreas, suggestive of a cystic neoplasm. The patient underwent an open distal pancreatectomy with splenectomy, and histopathological analysis confirmed the diagnosis of SPN of the pancreas. CONCLUSION This case highlights the risk of SPN in adolescent girls and the necessity of early diagnosis and intervention for better outcomes.
Collapse
Affiliation(s)
- Aakriti Sapkota
- Department of Medicine, B.P. Koirala Institute of Health Sciences, Dharan 56700, Koshi, Nepal
| | - Rajesh Paudel
- Department of Radiodiagnosis and Imaging, B.P. Koirala Institute of Health Sciences, Dharan 56700, Koshi, Nepal
| | - Sandip Pandey
- Department of Medicine, B.P. Koirala Institute of Health Sciences, Dharan 56700, Koshi, Nepal
| | - Navin Bhatt
- Department of Medicine, NYC Health + Hospitals/Elmhurst, Icahn School of Medicine at Mount Sinai, Elmhurst, NY 11373, United States
| |
Collapse
|
46
|
Song L, Wang G, Chen Z, Wang G. Research status of risk factors and prevention of pancreatic cancer: A bibliometric and visual analysis. Medicine (Baltimore) 2025; 104:e41831. [PMID: 40101086 PMCID: PMC11922469 DOI: 10.1097/md.0000000000041831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
One of the biggest public health issues facing the globe today is pancreatic cancer (PC). To serve as a guide for clinically identifying existing research hotspots and conducting related studies in the future, bibliometric and visualization analyses of the literature on risk factors and PC prevention were carried out in this work. Results of published research from 2004 to 2024 were retrieved using the Web of Science database as a search platform. CiteSpace and VOSviewer were used for bibliometric and visual analysis. Based on the exclusion criteria, 868 articles in all were screened. Between 2004 and 2024, the quantity of articles published varied. Between 2017 and 2023, there was a consistent upward trend in the quantity of published literature. Cancer epidemiology biomarkers and prevention, cancers, and the Asian Pacific Journal of Cancer Prevention were the 3 journals with the most publications. The 2 nations with the most publications are China and the United States. The 2 nations with the most publications are China and the United States. The top 3 most published universities are Harvard University, the National Institutes of Health (NIH), and the National Cancer Institute (NCI). Buzzwords include body mass index, obesity, diabetes, smoking, and exercise.
Collapse
Affiliation(s)
- Lichen Song
- School of Clinical Medicine, Dali University, Dali, China
| | - Guihua Wang
- Respiratory and Critical Care Medicine Department, The First Affiliated Hospital of Dali University, Dali, China
| | - Ziyi Chen
- School of Clinical Medicine, Dali University, Dali, China
| | - Guangming Wang
- Center of Genetic Testing, The First Affiliated Hospital of Dali University, Dali, China
| |
Collapse
|
47
|
Cox M, Vitello D, Chawla A. Translating the multifaceted use of liquid biopsy to management of early disease in pancreatic adenocarcinoma. Front Oncol 2025; 15:1520717. [PMID: 40182037 PMCID: PMC11966063 DOI: 10.3389/fonc.2025.1520717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related mortality, primarily due to late stage at diagnosis. This review examines the multifaceted applications of liquid biopsy and circulating tumor DNA (ctDNA) analysis in the diagnosis and management of PDAC. We review the current literature on the technological advancements in liquid biopsy analysis such as next generation sequencing (NGS) and digital droplet PCR (ddPCR) as well as multi-omics technologies, highlighting their potential for accurate molecular subtyping through ctDNA analysis. This review highlights the significant role of ctDNA in the assessment of tumor behavior, disease subtyping, prediction and monitoring of treatment response, and evaluation of minimal residual disease. We discuss the implications of integrating liquid biopsy techniques into clinical practice as well as its challenges and limitations. By drawing insights from recent studies, this review aims to provide a comprehensive overview of how liquid biopsy and ctDNA analysis can enhance early disease management strategies in PDAC. We underscore the need for additional prospective studies and clinical trials to validate its feasibility and accuracy in order to establish clinical utility, with the ultimate goal of routine incorporation into practice to improve patient outcomes and transform the treatment landscape for PDAC.
Collapse
Affiliation(s)
- Madison Cox
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, United States
| | - Dominic Vitello
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Akhil Chawla
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, United States
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, United States
| |
Collapse
|
48
|
Yin J, Li J, Wang H. Disulfidptosis: a novel gene-based signature predicts prognosis and immunotherapy efficacy of pancreatic adenocarcinoma. Discov Oncol 2025; 16:308. [PMID: 40072658 PMCID: PMC11904034 DOI: 10.1007/s12672-025-02053-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Disulfidptosis, a novel form of disulfide stress-induced cell death involved in tumor progression, hasn't be well defined the function in tumor progression. And the clinical impacts of disulfidptosis-related genes (DRGs) in pancreatic adenocarcinoma (PAAD) remain largely unclear. In this study, we identified two distinct disulfidptosis subtypes and found that multilayer DRG alterations were associated with prognosis and TME infiltration characteristics. A three-gene prognostic signature was constructed to predict prognosis, and its clinical significance was characterized in the TCGA-PAAD cohort. The disulfidptosis signature was significantly correlated with prognosis, molecular subtype, CD8 T-cell infiltration, response to immune checkpoint inhibitors and chemotherapeutic drug sensitivity, and its predictive capability in PAAD patients was validated in multiple cohorts. Meanwhile, two anti-PD-L1 immunotherapy cohorts confirmed that low-risk patients exhibited substantially enhanced clinical response and treatment advantages. Furthermore, the expression patterns of DRGs were validated by quantitative real-time PCR. The expression and prognostic predictive capability of GLUT1 were verified by 87 PAAD patients from our cohort. These findings may help us understand the roles of DRGs in PAAD and the molecular characterization of disulfidptosis subtypes. The disulfidptosis signature could be a promising biomarker for prognosis, molecular subtypes, TME infiltration characteristics and immunotherapy efficacy.
Collapse
Affiliation(s)
- Jingyang Yin
- Department of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, China
- University of Chinese Academy of Sciences (UCAS) Chongqing School, Chongqing Medical University, Chongqing, China
| | - Jian Li
- Department of Gastrointestinal Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710000, China
| | - Huaizhi Wang
- Department of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, China.
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, China.
- University of Chinese Academy of Sciences (UCAS) Chongqing School, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
49
|
Cullinane C, Devine M, Alazzawi M, Suilleabhain CO, Sullivan AO. "Somatostatin analogues do not reduce the risk of clinically relevant post-operative fistula rates in patients undergoing pancreatic surgery", a systematic review and meta-analysis. HPB (Oxford) 2025:S1365-182X(25)00078-4. [PMID: 40180812 DOI: 10.1016/j.hpb.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND The impact of Somatostatin Analogues (SSA) on Post-Operative Pancreatic Fistula (POPF) risk reduction lacks clarity and reports are conflicting. The aim of this study was to perform a systematic review and meta-analysis to explore the effect of SSA on POPF rates. METHODS A systematic review was performed for studies reporting POPF in relation to SSA use following the consensus by the International Study Group of Pancreatic Fistula (ISGPF) to re-define what constitutes a clinically significant POPF in 2015. The primary outcome was the incidence of clinically relevant POPF among patients who received SSA peri-operatively. RESULTS Twenty studies, including 6947 patients, were eligible for inclusion. Overall, SSA use did not significantly lower the risk of developing a POPF(OR 0.89, 95 % CI 0.66-1.20, P = 0.44, I2 = 73 %). Subgroup analysis was performed to determine whether SSA could reduce POPF in high-risk cohorts (soft pancreas, duct <5 mm). SSA did not significantly reduce POPF in the five studies reporting on high-risk cohorts (OR 1.42, 95 % CI 0.60-3.37, P = 0.43, I2 = 73 %). Furthermore, subgroup analysis of both grade B and grade C POPF's did not show any benefit of SSA. CONCLUSION SSA prophylaxis does not reduce the incidence of clinically relevant POPR and should not be routinely administered for pancreatic resections.
Collapse
Affiliation(s)
- Carolyn Cullinane
- Department of Hepatobiliary and Pancreatic Surgery, Mercy University Hospital, Cork, Ireland; Department of General Surgery, Royal College of Surgeons, Dublin, Ireland.
| | - Michael Devine
- Department of General Surgery, Royal College of Surgeons, Dublin, Ireland
| | - Mohammed Alazzawi
- Department of General Surgery, Royal College of Surgeons, Dublin, Ireland
| | | | - Adrian O Sullivan
- Department of Hepatobiliary and Pancreatic Surgery, Mercy University Hospital, Cork, Ireland
| |
Collapse
|
50
|
Zhang B, Lang Z, Zhu K, Luo W, Zhao Z, Zhang Z, Wang Z. Whether preoperative biliary drainage leads to better patient outcomes of pancreaticoduodenectomy: a meta-analysis and systematic review. BMC Gastroenterol 2025; 25:161. [PMID: 40069629 PMCID: PMC11900024 DOI: 10.1186/s12876-025-03761-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
OBJECTIVE To investigate whether preoperative biliary drainage is beneficial for patients undergoing pancreaticoduodenectomy. METHODS The PubMed, Cochrane Library and the Web of Science were systematically searched for relevant trials that included outcome of pancreaticoduodenectomy with and without preoperative biliary drainage from January 2010 to May 2024. The primary outcomes are postoperative pancreatic fistula and intra-abdominal infection. Data is pooled using the risk ratio or standardized mean difference with 95% confidence interval. The study protocol was registered prospectively with PROSPERO (CRD42022372584). RESULTS A total of 39 retrospective cohort studies with 33,516 patients were included in this trial. Compared with no preoperative biliary drainage, the preoperative biliary drainage group had a longer hospital stay (SMD, 0.14). Performing preoperative biliary drainage significantly increases the risk of postoperative pancreatic fistula (RR, 1.09), intra-abdominal infection (RR, 1.09), surgical site infection (RR, 1.84), and sepsis (RR, 1.37). But preoperative biliary drainage lowers risk of bile leak (RR, 0.74). CONCLUSION Preoperative biliary drainage before pancreaticoduodenectomy increases the risk of postoperative complications without clear overall benefits. Routine PBD is not recommended for younger patients with mild to moderate jaundice but may be considered for high-risk patients, such as those with severe infections or progressive jaundice. Optimizing preoperative biliary drainage duration and timing may help reduce complications. Further research is needed to refine patient selection and perioperative strategies.
Collapse
Affiliation(s)
- Bo Zhang
- The Forth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Zekun Lang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Kexiang Zhu
- The Forth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Wei Luo
- The Forth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Zhenjie Zhao
- The Forth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Zeliang Zhang
- The Forth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Zhengfeng Wang
- The Forth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China.
| |
Collapse
|