101
|
Heller M, Mann DA, Katona BW. Current Approaches of Pancreatic Cancer Surveillance in High-Risk Individuals. J Gastrointest Cancer 2025; 56:61. [PMID: 39932614 PMCID: PMC11814005 DOI: 10.1007/s12029-025-01184-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2025] [Indexed: 02/14/2025]
Abstract
Currently, those recommended to undergo pancreatic cancer (PC) surveillance include appropriately aged individuals at high risk of PC due to an identifiable genetic susceptibility or those without identifiable genetic susceptibility who nonetheless have a strong family history of PC. With increases in identification of individuals at high risk for PC and increased use of PC surveillance in clinical practice, there has been increasing debate about who should undergo surveillance as well as how surveillance should be performed including use of imaging and blood-based testing. Furthermore, there is increasing interest in the outcomes of PC surveillance in high-risk individuals with some studies demonstrating that surveillance leads to downstaging of PC and improvements in survival. In this review, we summarize the current state of PC surveillance in high-risk individuals, providing an overview of the risk factors associated with PC, selection of high-risk individuals for PC surveillance, and the current, but non-uniform, recommendations for performing PC surveillance. Additionally, we review approaches to apply various imaging and blood-based tests to surveillance and the outcomes of PC surveillance.
Collapse
Affiliation(s)
- Melissa Heller
- Division of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Derek A Mann
- Division of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bryson W Katona
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd., 751 South Pavilion, Philadelphia, PA, 19104, USA.
| |
Collapse
|
102
|
Hayat U, Croce PS, Saadeh A, Desai K, Appiah J, Khan S, Khan YI, Kumar K, Hanif A. Current and Emerging Treatment Options for Pancreatic Cancer: A Comprehensive Review. J Clin Med 2025; 14:1129. [PMID: 40004658 PMCID: PMC11856716 DOI: 10.3390/jcm14041129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of death worldwide, and its global burden has increased significantly over the past few years. The incidence of pancreatic cancer has also increased in the United States, and most of this increase is attributed to the population's aging process in addition to the rise in the prevalence of risk factors such as obesity, diabetes, smoking, and alcohol intake. Most patients with pancreatic cancer present with advanced unresectable or metastatic disease. Only a few patients present at an early stage with localized disease, and a multidisciplinary approach is required to maximize survival and outcomes. The surgical approach is an option for localized disease, and surgery's safety and efficacy have also been improved in recent years due to the increasing use of minimally invasive surgical techniques. Moreover, systematic chemotherapy has also been used and has had a significant impact on survival. More recently, neoadjuvant therapy has been used for pancreatic cancer along with radiation therapy, optimizing survival among those patients. Targeted therapies have been introduced based on genetic testing in metastatic pancreatic cancer and have shown promising results. Moreover, immune checkpoint inhibitors and targeted agents such as PARP inhibitors and vaccines have emerged with optimal results in terms of survival. To conclude, pancreatic cancer is considered a disease with poor long-term survival; however, recent developments in pharmacotherapy have changed its treatment and have improved outcomes with improved survival. Our review summarizes ongoing therapeutic options for local and metastatic pancreatic cancer. It also summarizes new state-of-the-art therapies that have emerged or are in trials, which can change the pancreatic cancer treatment perspective.
Collapse
Affiliation(s)
- Umar Hayat
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA; (P.S.C.); (K.D.); (J.A.); (S.K.)
| | - Phillip S. Croce
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA; (P.S.C.); (K.D.); (J.A.); (S.K.)
| | - Aseel Saadeh
- Department of Internal Medicine, Geisinger Medical Center, Danville, PA 18711, USA;
| | - Karna Desai
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA; (P.S.C.); (K.D.); (J.A.); (S.K.)
| | - John Appiah
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA; (P.S.C.); (K.D.); (J.A.); (S.K.)
| | - Sidrah Khan
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA; (P.S.C.); (K.D.); (J.A.); (S.K.)
| | - Yakub I. Khan
- Department of Internal Medicine, Division of Gastroenterology, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA; (Y.I.K.); (K.K.)
| | - Kishore Kumar
- Department of Internal Medicine, Division of Gastroenterology, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA; (Y.I.K.); (K.K.)
| | - Ahmad Hanif
- Department of Internal Medicine, Division of Hematology/Oncology, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, PA 18711, USA;
| |
Collapse
|
103
|
Lin Y, Liao Y, Shen J. Angiotensin system inhibitors improve survival in patients undergoing pancreatic cancer resection: a meta-analysis of real-world evidence. Expert Rev Anticancer Ther 2025:1-8. [PMID: 39910780 DOI: 10.1080/14737140.2025.2464208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/15/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND The role of angiotensin system inhibitors (ASIs) in modifying the prognosis for patients undergoing pancreatic cancer resection is not yet definitively established. This meta-analysis endeavors to consolidate existing real-world data to provide a robust, evidence-based assessment of their impact on clinical outcomes. METHODS A meticulous search strategy was devised and executed across PubMed, Embase, and Web of Science databases to retrieve all relevant studies evaluating the prognostic impact of ASIs in patients who have undergone resection for pancreatic cancer. Studies comparing survival outcomes between ASI users and non-users were included in the meta-analysis. Publication bias was assessed using funnel plotand Egger's test. Sensitivity analysis employing the leave-one-out approach was conducted to ensure the robustness and reliability of the pooled estimate. RESULTS Seven studies encompassing 8,549 patients were analyzed. The utilization of ASIs was significantly associated with improved overall survival (HR: 0.78; 95%CI: 0.68-0.89) in patients undergoing pancreatic cancer resection. Sensitivity analysis further validated the consistency and stability of the pooled result. CONCLUSION Current clinical evidence suggests that ASIs are associated with improved prognosis in patients who have undergone pancreatic cancer resection. These findings highlight the potential of ASIs as a beneficial adjunctive therapy in the management of resected pancreatic cancer, warranting their consideration in clinical management protocols. REGISTRATION PROSPERO (identifier: CRD42024580624).
Collapse
Affiliation(s)
- Yuxuan Lin
- Department of Pharmacy, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, Guangxi, PR China
| | - Yonghe Liao
- College of Pharmaceutical Science, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Jinhai Shen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, PR China
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, Jiangsu, PR China
| |
Collapse
|
104
|
Phan TAT, Ngo KKH, Nguyen TCT, Mai TT, Nguyen HD, Duong TT, Tran LP, Duong TT, Huynh TKC, Koroleva EV, Ignatovich ZV, Ermolinskaya AL, Nguyen HP, Nguyen THA, Ton AK, Do TH, Hoang TKD. 2-Amino-4,6-diarylpyrimidines as potential chronic myeloid leukemia cell inhibitors targeting anti-ABL1 kinase: microwave-assisted synthesis, biological evaluation, molecular docking, and dynamics studies. RSC Adv 2025; 15:4458-4471. [PMID: 39931407 PMCID: PMC11808659 DOI: 10.1039/d4ra08330j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
In this work, a simple and mild process was used to synthesize a series of 2-amino-4,6-diarylpyrimidine derivatives, 1a-1q, whose structures were verified by FTIR, 1D- and 2D-NMR, and HRMS techniques, to investigate and develop anticancer agents. Under microwave irradiation, a two-step process was carried out, consisting of aldol condensation of benzaldehydes and acetophenones to produce intermediate chalcones and ring closure condensation of chalcones and guanidine hydrochloride. Each generated compound's anticancer activity against the human chronic myelocytic leukemia K562 cancer cell line was investigated in vitro to determine the active compounds, which were subsequently evaluated for inhibiting the ABL1 tyrosine kinase. According to these findings, compound 1e demonstrated considerable inhibition against K562 cancer cells and ABL1 tyrosine kinase at IC50 values of 8.77 ± 0.55 μM and 3.35 ± 0.58 μM, respectively. The molecular docking on wild-type and mutant type ABL1 (PDB ID 2HYY and 5MO4) investigation indicated that 1e and 1g interacted with amino acids. It formed stable hydrogen bonds and π-π linkages with crucial residues in the active site of the enzyme. Moreover, the stability of these enzyme-ligand complexes was confirmed using molecular dynamics simulations. These findings suggested that compounds 1e and 1g can be considered promising cancer treatment agents.
Collapse
Affiliation(s)
- Thi-Anh-Truc Phan
- Institute of Chemical Technology, Vietnam Academy of Science and Technology No. 1A, TL29 St., Thanh Loc Ward, Dist. 12 Ho Chi Minh City 70000 Vietnam
- Ton Duc Thang University No. 19, Nguyen Huu Tho St., Tan Hung Ward, Dist. 7 Ho Chi Minh City 70000 Vietnam
| | - Kim-Khanh-Huy Ngo
- Institute of Chemical Technology, Vietnam Academy of Science and Technology No. 1A, TL29 St., Thanh Loc Ward, Dist. 12 Ho Chi Minh City 70000 Vietnam
| | - Thi-Cam-Thu Nguyen
- Institute of Chemical Technology, Vietnam Academy of Science and Technology No. 1A, TL29 St., Thanh Loc Ward, Dist. 12 Ho Chi Minh City 70000 Vietnam
| | - Thanh-Tan Mai
- University of Medicine and Pharmacy at Ho Chi Minh City No. 41, Dinh Tien Hoang St., Ben Nghe Ward, Dist. 1 Ho Chi Minh City 70000 Vietnam
| | - Hai-Dang Nguyen
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology No. 18, Hoang Quoc Viet St., Nghia Do Ward, Cau Giay District Ha Noi City 100000 Vietnam
| | - Thu-Trang Duong
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology No. 18, Hoang Quoc Viet St., Nghia Do Ward, Cau Giay District Ha Noi City 100000 Vietnam
| | - Le-Phu Tran
- Institute of Chemical Technology, Vietnam Academy of Science and Technology No. 1A, TL29 St., Thanh Loc Ward, Dist. 12 Ho Chi Minh City 70000 Vietnam
- Ton Duc Thang University No. 19, Nguyen Huu Tho St., Tan Hung Ward, Dist. 7 Ho Chi Minh City 70000 Vietnam
| | - Thanh-Tuyen Duong
- Institute of Chemical Technology, Vietnam Academy of Science and Technology No. 1A, TL29 St., Thanh Loc Ward, Dist. 12 Ho Chi Minh City 70000 Vietnam
- Ton Duc Thang University No. 19, Nguyen Huu Tho St., Tan Hung Ward, Dist. 7 Ho Chi Minh City 70000 Vietnam
| | - Thi-Kim-Chi Huynh
- Institute of Chemical Technology, Vietnam Academy of Science and Technology No. 1A, TL29 St., Thanh Loc Ward, Dist. 12 Ho Chi Minh City 70000 Vietnam
| | - Elena V Koroleva
- Institute of Chemistry of New Materials, National Academy of Sciences of Belarus 36, F. Skorina St. Minsk 220141 Republic of Belarus
| | - Zhanna V Ignatovich
- Institute of Chemistry of New Materials, National Academy of Sciences of Belarus 36, F. Skorina St. Minsk 220141 Republic of Belarus
| | - Anastasiya L Ermolinskaya
- Institute of Chemistry of New Materials, National Academy of Sciences of Belarus 36, F. Skorina St. Minsk 220141 Republic of Belarus
| | - Hoang-Phuc Nguyen
- Institute of Chemical Technology, Vietnam Academy of Science and Technology No. 1A, TL29 St., Thanh Loc Ward, Dist. 12 Ho Chi Minh City 70000 Vietnam
| | - Thi-Hong-An Nguyen
- Institute of Chemical Technology, Vietnam Academy of Science and Technology No. 1A, TL29 St., Thanh Loc Ward, Dist. 12 Ho Chi Minh City 70000 Vietnam
| | - Anh-Khoa Ton
- Institute of Chemical Technology, Vietnam Academy of Science and Technology No. 1A, TL29 St., Thanh Loc Ward, Dist. 12 Ho Chi Minh City 70000 Vietnam
| | - Tuong-Ha Do
- Ton Duc Thang University No. 19, Nguyen Huu Tho St., Tan Hung Ward, Dist. 7 Ho Chi Minh City 70000 Vietnam
| | - Thi-Kim-Dung Hoang
- Institute of Chemical Technology, Vietnam Academy of Science and Technology No. 1A, TL29 St., Thanh Loc Ward, Dist. 12 Ho Chi Minh City 70000 Vietnam
| |
Collapse
|
105
|
Shi R, Zhang N, Li H, Zhong H, Zhong C, Du W, Yang X. Cancer-associated fibroblast-derived COL17A1 promotes gemcitabine resistance and tumorigenesis in pancreatic cancer cells by interacting with ACTN4. Discov Oncol 2025; 16:118. [PMID: 39907925 PMCID: PMC11799490 DOI: 10.1007/s12672-025-01825-8] [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: 08/28/2024] [Accepted: 01/16/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are key components of tumor microenvironment and have been identified to be involved in modulating drug resistance in cancers by secreting molecules. Pancreatic cancer (PC) is a leading cause of cancer mortality with high aggressiveness. Gemcitabine (GEM) is one of primary antineoplastic drugs for PC. Collagen XVII (COL17A1) expression was found to be upregulated in GEM-resistant CAFs. Here, this study focused on investigating whether CAFs affected GEM resistance in PC by secreting COL17A1 and its associated mechanisms. METHODS In total, 60 newly diagnosed PC patients only with GEM-based chemotherapy were recruited. Normal fibroblasts (NFs) and CAFs were isolated using fresh normal and resistant PC tissues. Human pancreatic duct epithelial (HPDE) cells were used for functional analyses. Levels of COL17A1 and Actinin Alpha 4 (ACTN4) were measured by using qRT-PCR and western blotting. Functional analyses were conducted using MTT, 5-ethynyl-2'-deoxyuridine, transwell, and sphere formation assays, respectively. The interaction between COL17A1 and ACTN4 was analyzed by Co-immunoprecipitation and immunofluorescence assays. Animal models were established for in vivo analysis. RESULTS CAF incubation promoted GEM resistance and enhanced the proliferation, invasion and stemness in GEM-resistant PC cells. COL17A1 was highly expressed in resistant CAFs and GEM-resistant PC cells, and CAF incubation could increase COL17A1 expression in resistant PC cells. Moreover, COL17A1 silencing in GEM-resistant PC cells or the incubation of COL17A1-decreased CAF with GEM-resistant PC cells could suppress GEM resistance and cell oncogenic phenotype progression. Mechanistically, COL17A1 interacted with ACTN4 protein, and the anticancer effects mediated by COL17A1-decreased CAFs in resistant PC cells were reversed by ACTN4 overexpression. In vivo assay also showed that COL17A1-decreased CAFs suppressed the growth and GEM resistance in PC by ACTN4. CONCLUSION CAFs-derived COL17A1 promoted GEM resistance and tumorigenesis in PC by interacting with ACTN4, suggesting a new method for overcoming GEM resistance in PC.
Collapse
Affiliation(s)
- Rongyu Shi
- Department of Hepatobiliary and Pancreatic Surgery, First People's Hospital of Jiashan County, Jiaxing, 314100, China
| | - Ning Zhang
- Department of Hepatobiliary and Pancreatic Surgery, First People's Hospital of Jiashan County, Jiaxing, 314100, China
| | - Han Li
- Department of Hepatobiliary and Pancreatic Surgery, First People's Hospital of Jiashan County, Jiaxing, 314100, China
| | - Hu Zhong
- Department of General Surgery, The Affiliated Shuyang Hospital of Xuzhou Medical University, No. 9, Yingbin Road, Suqian, 223600, China
| | - Chengcheng Zhong
- Department of General Surgery, The Affiliated Shuyang Hospital of Xuzhou Medical University, No. 9, Yingbin Road, Suqian, 223600, China
| | - Wei Du
- Department of Hepatobiliary and Pancreatic Surgery, First People's Hospital of Jiashan County, Jiaxing, 314100, China
| | - Xi Yang
- Department of General Surgery, The Affiliated Shuyang Hospital of Xuzhou Medical University, No. 9, Yingbin Road, Suqian, 223600, China.
| |
Collapse
|
106
|
Zhang N, Chen S, Shi Y, Wang Z, Jia R, Dai G. Charlson syndrome index predicted survival in pancreatic cancer patients received immunotherapy. Front Immunol 2025; 16:1487318. [PMID: 39963141 PMCID: PMC11831428 DOI: 10.3389/fimmu.2025.1487318] [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: 08/27/2024] [Accepted: 01/03/2025] [Indexed: 02/20/2025] Open
Abstract
Objective The Charlson Comorbidity Index (CCI) is a widely utilized assessment tool for evaluating the mortality rate among patients with chronic diseases and tumors. Currently, there is a dearth of research investigating the correlation between CCI and survival rates in advanced pancreatic cancer patients received immunotherapy. Therefore, this study aims to elucidate the association between CCI and survival rates in real-world settings for pancreatic cancer patients received immunotherapy. Methods A total of 104 patients with advanced pancreatic cancer who received immunotherapy at the General Hospital of the People's Liberation Army between September 2015 and September 2020 were included in this study. The patients were categorized into two groups based on their Charlson Comorbidity Index (CCI) scores: low CCI group (CCI <7) and high CCI group (CCI ≥7). The statistical analysis focused on examining the correlation between CCI score and survival outcome. Results The high CCI group exhibited significantly lower overall survival (OS) and progression-free survival (PFS) compared to the low CCI group (p<0.05). The median OS for the high CCI and low CCI groups were 7.82 and 44.17 months, respectively, while the median PFS were 2.40 and 6.40 months, respectively. Multivariate analysis revealed that high CCI was independently risk factor for both OS (HR=2.801, 95%CI: 1.433-5.472, p=0.003) and PFS (HR=2.546, 95%CI: 1.389-4.668, p=0.003). Conclusion The CCI score serves as a significant independent predictive indicator for advanced pancreatic cancer patients received immunotherapy.
Collapse
Affiliation(s)
- Nan Zhang
- Medical School of Chinese PLA, Beijing, China
- Department of Medical Oncology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shiyun Chen
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yue Shi
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhikuan Wang
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ru Jia
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Guanghai Dai
- Department of Medical Oncology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
107
|
Melle F, Menon D, Conniot J, Ostolaza-Paraiso J, Mercado S, Oliveira J, Chen X, Mendes BB, Conde J, Fairen-Jimenez D. Rational Design of Metal-Organic Frameworks for Pancreatic Cancer Therapy: from Machine Learning Screening to In Vivo Efficacy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2412757. [PMID: 39895194 DOI: 10.1002/adma.202412757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/09/2024] [Indexed: 02/04/2025]
Abstract
Despite improvements in cancer survival rates, metastatic and surgery-resistant cancers, such as pancreatic cancer, remain challenging, with poor prognoses and limited treatment options. Enhancing drug bioavailability in tumors, while minimizing off-target effects, is crucial. Metal-organic frameworks (MOFs) have emerged as promising drug delivery vehicles owing to their high loading capacity, biocompatibility, and functional tunability. However, the vast chemical diversity of MOFs complicates the rational design of biocompatible materials. This study employed machine learning and molecular simulations to identify MOFs suitable for encapsulating gemcitabine, paclitaxel, and SN-38, and identified PCN-222 as an optimal candidate. Following drug loading, MOF formulations are improved for colloidal stability and biocompatibility. In vitro studies on pancreatic cancer cell lines have shown high biocompatibility, cellular internalization, and delayed drug release. Long-term stability tests demonstrated a consistent performance over 12 months. In vivo studies in pancreatic tumor-bearing mice revealed that paclitaxel-loaded PCN-222, particularly with a hydrogel for local administration, significantly reduced metastatic spread and tumor growth compared to the free drug. These findings underscore the potential of PCN-222 as an effective drug delivery system for the treatment of hard-to-treat cancers.
Collapse
Affiliation(s)
- Francesca Melle
- The Adsorption & Advanced Materials Laboratory (AAML), Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Dhruv Menon
- The Adsorption & Advanced Materials Laboratory (AAML), Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - João Conniot
- NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC), NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Jon Ostolaza-Paraiso
- The Adsorption & Advanced Materials Laboratory (AAML), Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Sergio Mercado
- The Adsorption & Advanced Materials Laboratory (AAML), Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Jhenifer Oliveira
- NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC), NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Xu Chen
- The Adsorption & Advanced Materials Laboratory (AAML), Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Bárbara B Mendes
- NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC), NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - João Conde
- NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC), NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - David Fairen-Jimenez
- The Adsorption & Advanced Materials Laboratory (AAML), Department of Chemical Engineering & Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| |
Collapse
|
108
|
Guo Y, Dou Y, Ma X, Li Z, Li H, Sun X, Gao C, Liang Y, Zhao T. Prognostic Significance of C-Reactive Protein or Prealbumin in Pancreatic Ductal Adenocarcinoma. J Surg Res 2025; 306:543-553. [PMID: 39889315 DOI: 10.1016/j.jss.2024.12.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 02/02/2025]
Abstract
INTRODUCTION To clarify the prognostic significance of the C-reactive protein to prealbumin ratio (CRP or PALB) in patients with pancreatic cancer after radical resection. METHODS A total of 432 patients with pathologically confirmed pancreatic ductal adenocarcinoma were enrolled in this retrospective study. The predictive capacity of various inflammatory indices was analyzed and compared using the area under the time-dependent receiver operating characteristic curve, including CRP or PALB, CRP-to-albumin ratio, neutrophil-to-lymphocyte ratio, and platelet-to-lymphocyte ratio. The univariate and multivariate Cox hazard models were employed to analyze the effects of CRP or PALB on overall survival (OS) and recurrence-free survival (RFS). RESULTS The optimal cut-off value for preoperative CRP or PALB was 5.94, which was derived from the receiver operating characteristic curve. In comparison with traditional inflammatory indices, CRP or PALB had the highest area under the time-dependent receiver operating characteristic curve (0.693 for 3-y OS, 0.664 for 3-y RFS, 0.662 for 5-y OS, and 0.670 for 5-y RFS), all with P < 0.05. However, when compared with neutrophil-to-lymphocyte ratio, the predictive power of CRP or PALB was not significant for 3-y RFS (P = 0.085). Based on the results of the univariate and multivariate survival analyses, patients in the high CRP or PALB group (HCP: CRP or PALB >5.94) exhibited significantly poorer OS and RFS (median OS: 20.0 versus. 38.0 mo, P = 0.003; median RFS: 10.0 versus. 22.0 mo, P < 0.001) than those in the low CRP or PALB group (CRP or PALB ≤5.94). The multivariate analysis indicated that the HCP was independently associated with poor OS (hazard ratio (HR): 1.556, 95% confidence interval (CI) [1.089-2.222], P = 0.015) and RFS (HR: 1.551, 95% CI [1.135-2.119], P = 0.006). CONCLUSIONS The predictive capacity of preoperative CRP or PALB in pancreatic ductal adenocarcinoma patients exceeds that of traditional inflammatory indices. HCP levels are significantly correlated with a poor prognosis.
Collapse
Affiliation(s)
- Yu Guo
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Yibo Dou
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Xi Ma
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Zhifei Li
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China; Department of Hepatobiliary-Pancreatic-Splenic Surgery, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Haorui Li
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Xugang Sun
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Chuntao Gao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Yuexiang Liang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China.
| | - Tiansuo Zhao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China.
| |
Collapse
|
109
|
Zhang Z, Guo S, Su W, Pan G, Cao K, Jiang H, Zhang L, Cheng C, Jin G, Zuo C. Preoperative assessment of pancreatic cancer with [ 68Ga]Ga-DOTA-FAPI-04 PET/MR versus [ 18F]-FDG PET/CT plus contrast-enhanced CT: a prospective preliminary study. Eur J Nucl Med Mol Imaging 2025; 52:1017-1027. [PMID: 39508900 DOI: 10.1007/s00259-024-06943-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/05/2024] [Indexed: 11/15/2024]
Abstract
PURPOSE To assess the diagnostic performance of [68Ga]Ga-DOTA-FAPI-04 PET/MR imaging in the preoperative evaluation of pancreatic cancer and compare it with that of [18F]-FDG PET/CT plus contrast-enhanced CT (CECT). METHODS Thirty-one patients with pancreatic cancer underwent preoperative [68Ga]Ga-DOTA-FAPI-04 PET/MR, [18F]-FDG PET/CT, and CECT imaging. Two nuclear medicine physicians independently reviewed two sets of images (set 1, [68Ga]Ga-DOTA-FAPI-04 PET/MR; set 2, [18F]-FDG PET/CT plus CECT) and reached a consensus on tumour resectability, N staging (N0 or N positive) and M staging (M0 or M1). Based on the above indices, the resectability of the tumour was determined according to a five-point scale. Clinical, operative, and pathological findings were used as a reference standard to compare the diagnostic performance of the two imaging sets via the McNemar test. RESULTS The diagnostic performance of [68Ga]Ga-DOTA-FAPI-04 PET/MR imaging was not significantly different from that of [18F]-FDG PET/CT plus CECT imaging in the assessment of tumour resectability (area under the receiver operating characteristic curve: 0.854 vs. 0.775, p = 0.192), N staging [accuracy: 82.4% (14 of 17 patients) vs. 58.8% (10 of 17 patients), p = 0.125] and M staging [accuracy: 100% (31 of 31 patients) vs. 90.3% (28 of 31 patients), p = 0.250]. However, compared with [18F]-FDG PET/CT plus CECT imaging, [68Ga]Ga-DOTA-FAPI-04 PET/MR imaging changed the M stage in three patients by upstaging from M0 to M1 in 2 patients and downstaging from M1 to M0 in 2 patients. In 13 patients with liver metastases, the number of liver metastases detected via [68Ga]Ga-DOTA-FAPI-04 PET/MR imaging was greater than that detected via [18F]-FDG PET/CT plus CECT imaging (324 vs. 240). In 3 patients with peritoneal metastases, [68Ga]Ga-DOTA-FAPI-04 PET/MR imaging detected more peritoneal metastases than did [18F]-FDG PET/CT plus CECT imaging. CONCLUSIONS [68Ga]Ga-DOTA-FAPI-04 PET/MR imaging has diagnostic accuracy comparable to [18F]-FDG PET/CT plus CECT in terms of preoperative staging and assessment of resectability in pancreatic cancer; additionally, it exhibits superior capability in detecting liver and peritoneal metastases. Consequently, [68Ga]Ga-DOTA-FAPI-04 PET/MR has the potential to become a one-stop imaging tool for the preoperative evaluation of pancreatic cancer.
Collapse
Affiliation(s)
- Zeyu Zhang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Yang Pu District, Shanghai, 200433, China
| | - Shiwei Guo
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Yang Pu District, Shanghai, 200433, China
| | - Weiwei Su
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Yang Pu District, Shanghai, 200433, China
- Department of Radiology, Naval Medical Centre of People's Liberation Army, Naval Medical University, Shanghai, 200050, China
| | - Guixia Pan
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Yang Pu District, Shanghai, 200433, China
| | - Kai Cao
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| | - Lu Zhang
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Yang Pu District, Shanghai, 200433, China
| | - Chao Cheng
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Yang Pu District, Shanghai, 200433, China.
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Yang Pu District, Shanghai, 200433, China.
| | - Changjing Zuo
- Department of Nuclear Medicine, Changhai Hospital, Naval Medical University, 168 Changhai Road, Yang Pu District, Shanghai, 200433, China.
| |
Collapse
|
110
|
Kurdyn A, Pawłowska M, Paluszkiewicz E, Cichorek M, Augustin E. c-Myc inhibition and p21 modulation contribute to unsymmetrical bisacridines-induced apoptosis and senescence in pancreatic cancer cells. Pharmacol Rep 2025; 77:182-209. [PMID: 39361216 PMCID: PMC11743403 DOI: 10.1007/s43440-024-00658-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 01/21/2025]
Abstract
BACKGROUND Pancreatic cancer (PC) is one of the most aggressive cancers and is the seventh leading cause of cancer-related death worldwide. PC is characterized by rapid progression and resistance to conventional treatments. Mutations in KRAS, CDKN2A, TP53, SMAD4/DPC4, and MYC are major genetic alterations associated with poor treatment outcomes in patients with PC. Therefore, optimizing PC therapy is a tremendous challenge. Unsymmetrical bisacridines (UAs), synthesized by our group, are new promising compounds that have exhibited high cytotoxicity and antitumor activity against several solid tumors, including pancreatic cancer. METHODS The cellular effects induced by UAs in PC cells were evaluated by MTT assay (cell growth inhibition), flow cytometry, and fluorescence and light microscopy (cell cycle distribution, apoptosis, and senescence detection). Analysis of the effects of UAs on the levels of proteins (c-Myc, p53, SMAD4, p21, and p16) was performed by Western blotting. RESULTS Apoptosis was the main triggered mechanism of death after UAs treatment, and induction of the SMAD4 protein can facilitate this process. c-Myc, which is one of the molecular targets of UAs, can participate in the induction of cell death in a p53-independent manner. Moreover, UAs can also induce accelerated senescence through the upregulation of p21. Notably, senescent cells can die via apoptosis after prolonged exposure to UAs. CONCLUSIONS UAs have emerged as potent anticancer agents that induce apoptosis by inhibiting c-Myc protein and triggering cellular senescence in a dose-dependent manner by increasing p21 levels. Thus, UAs exhibit desirable features as promising candidates for future pancreatic anticancer therapies.
Collapse
Affiliation(s)
- Agnieszka Kurdyn
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, Gdańsk, 80-233, Poland
| | - Monika Pawłowska
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, Gdańsk, 80-233, Poland
| | - Ewa Paluszkiewicz
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, Gdańsk, 80-233, Poland
| | - Mirosława Cichorek
- Department of Embryology, Medical University of Gdańsk, Dębinki 1, Gdańsk, 80-211, Poland
| | - Ewa Augustin
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, Gabriela Narutowicza 11/12, Gdańsk, 80-233, Poland.
| |
Collapse
|
111
|
Li P, Wang K, Song J, Chen Z, Li Y, Chen Z. THBS1 knockdown suppresses pancreatic cancer progression through JAK2/STAT3 signaling pathway. Mol Cell Probes 2025; 79:102003. [PMID: 39710065 DOI: 10.1016/j.mcp.2024.102003] [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: 10/14/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Thrombospondin 1 (THBS1), a secreted protein, is implicated in the progression of numerous cancers, yet its specific contributions to pancreatic cancer (PC) remain underexplored. METHODS The association between THBS1 levels and prognosis in PC was investigated. Functional experiments in vitro were used to determine the cell functions of siTHBS1 through CCK8 assay for cell proliferation, Muse® Cell Analyzer for apoptosis, and transwell assay for invasion and migration. Colivelin was applied in recovery experiment to investigate the mechanism of THBS1 regulating the JAK2/STAT3 pathway in BXPC-3 cell. In addition, the LV-shTHBS1 lentivirus was used to construct subcutaneous tumors in nude mice to verify the function of THBS1 in vivo. RESULTS THBS1 expression was elevated in PC and associated with a poorer prognosis. THBS1 was highly expressed in these PC cells. siTHBS1 repressed cell growth, migration and invasiveness, while promoting apoptosis of BXPC-3 cells. THBS1 suppression also led to a decrease in the phosphorylation of JAK2 and STAT3. JAK2/STAT3 signaling activator (Colivelin) could partially reverse the biological effects. In addition, shTHBS1 can suppress the growth of implanted tumors in nude mice. CONCLUSIONS THBS1 knockdown suppressed cell proliferation, migration, and invasion while enhanced cell apoptosis through the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Ping Li
- Digestive Endoscopy Center, Hainan Cancer Hospital, Haikou, Hainan, 570100, China
| | - Kaixuan Wang
- Department of Gastroenterology, Changhai Hospital, Naval Military Medical University, Shanghai, 200433, China
| | - Jian Song
- Department of Gastroenterology, Hainan Cancer Hospital, Haikou, Hainan, 570100, China.
| | - Zhuang Chen
- Department of Gastroenterology, Hainan Cancer Hospital, Haikou, Hainan, 570100, China
| | - Yongyu Li
- Department of Gastroenterology, Hainan Cancer Hospital, Haikou, Hainan, 570100, China
| | - Zhaowei Chen
- Digestive Endoscopy Center, Hainan Cancer Hospital, Haikou, Hainan, 570100, China
| |
Collapse
|
112
|
Li R, Ji Q, Fu S, Gu J, Liu D, Wang L, Yuan X, Wen Y, Dai C, Li H. ITGA3 promotes pancreatic cancer progression through HIF1α- and c-Myc-driven glycolysis in a collagen I-dependent autocrine manner. Cancer Gene Ther 2025; 32:240-253. [PMID: 39690180 DOI: 10.1038/s41417-024-00864-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024]
Abstract
Pancreatic cancer is characterized by severe metabolic stress due to its prominent desmoplasia and poor vascularization. Integrin subunit alpha 3 (ITGA3) is a cell surface adhesion protein involved in tumor progression. However, the role of ITGA3 in pancreatic cancer progression, especially in metabolic reprogramming, remains largely unknown. In this study, we found that ITGA3 expression is elevated in pancreatic cancer tissues and predicts poor prognosis for patients with pancreatic cancer. Functional assays revealed that ITGA3 promotes the growth and liver metastasis of pancreatic cancer via boosting glycolysis. Mechanistically, Collagen I (Col1) derived from cancer cells acts as a ligand for ITGA3 to activate the FAK/PI3K/AKT/mTOR signaling pathway in an autocrine manner, thereby increasing the expression of HIF1α and c-Myc, two critical regulators of glycolysis. Blockade of Col1 by siRNA or of ITGA3 by a blocking antibody leads to specific inactivation of the FAK/PI3K/AKT/mTOR pathway and impairs malignant tumor behaviors induced by ITGA3. Thus, our data indicate that ITGA3 enhances glycolysis to promote pancreatic cancer growth and metastasis via increasing HIF1α and c-Myc expression in a Col1-dependent autocrine manner, making ITGA3 as a candidate diagnostic biomarker and a potential therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Rongkun Li
- Chest Oncology Department, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Qian Ji
- Department of Pulmonary Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Shengqiao Fu
- Chest Oncology Department, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Jichun Gu
- Department of Pancreatic surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Dejun Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lu Wang
- Abdominal Oncology Department, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Xiao Yuan
- Chest Oncology Department, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yi Wen
- Chest Oncology Department, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Chunhua Dai
- Chest Oncology Department, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Hengchao Li
- Department of Pancreatic surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
113
|
Zhou X, Cai M, Yang F, Huang L, Ling Y, Zhang Y, Nie H, Xing R. Hypoxia-induced autophagy in pancreatic cancer counteracts the cytotoxicity of CD8 + T cells by inhibiting the expression of MHC-I. Genes Immun 2025; 26:45-53. [PMID: 39715814 DOI: 10.1038/s41435-024-00315-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 12/02/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
The hypoxic microenvironment is an essential feature of solid tumors. Autophagy has been controversial in its role in immune regulation. This project aims to elucidate the impact of autophagy in pancreatic cancer (PC) under specific conditions (hypoxia) on CD8+ T cells and the regulatory mechanisms behind it.The levels of HIF1α and autophagy were analyzed by western blot (WB) and immunofluorescence (IF). The effects of HIF1α on cell autophagy were assessed in normoxic or hypoxic treatments using KC7F2 (HIF-1 channel inhibitor) or chloroquine (autophagy inhibitor). CD8+ T cells were co-cultured with PC cells to assess the cytotoxicity using lactate dehydrogenase (LDH) and Hoechst/PI staining. The content of cytokines and the activation level of CD8+ T cells were measured by enzyme-linked immunosorbent assay (ELISA) and flow cytometry. MHC-I expression in PC cells (membranes) was analyzed using quantitative reverse transcription polymerase chain reaction (qRT-PCR), WB, IF, and flow cytometry. Humanized immune-reconstituted mice were applied to investigate the impact of HIF1α-induced autophagy on in vivo immunity.When cells were in hypoxia, the levels of HIF1α and autophagy were higher compared to normoxic conditions. Treatment with KC7F2 resulted in similar levels of HIF1α and autophagy as those in normoxic state. Chloroquine treatment reversed the autophagy level to the normoxic state. The autophagy level of PC cells transfected with oe-HIF1α was increased, with reduced MHC-I expression on cells (membranes), which impaired the cytotoxicity of CD8+ T cells, and thus decreasing the probability of recognition and attack by CD8+ T cells when co-cultured with them. In mice, overexpression of HIF1α hindered the immune suppressive function of CD8+ T cells and facilitated the immune escape of PC by reducing antigen presentation of MHC-I.Under hypoxia, HIF1α-induced autophagy reduces the cytotoxicity of CD8+ T cells by repressing MHC-I expression.
Collapse
Affiliation(s)
- Xianfei Zhou
- Department of Hepatobiliary Surgery, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Miaoguo Cai
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Fan Yang
- Department of Hepatobiliary Surgery, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Luoshun Huang
- Department of Hepatobiliary Surgery, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Yisheng Ling
- Department of Hepatobiliary Surgery, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Yang Zhang
- Department of Hepatobiliary Surgery, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Hanqiu Nie
- Department of Hepatobiliary Surgery, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Renwei Xing
- Department of Hepatobiliary Surgery, Municipal Hospital Affiliated to Taizhou University, Taizhou, China.
| |
Collapse
|
114
|
Xing FL, Li BR, Fang YJ, Liang C, Liu J, Wang W, Xu J, Yu XJ, Qin Y, Zhang B. G3BP2 promotes tumor progression and gemcitabine resistance in PDAC via regulating PDIA3-DKC1-hENT in a stress granules-dependent manner. Acta Pharmacol Sin 2025; 46:474-488. [PMID: 39289547 PMCID: PMC11746999 DOI: 10.1038/s41401-024-01387-5] [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: 12/06/2023] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is distinguished by its aggressive malignancy, limited treatment avenues and a tendency towards chemotherapy resistance, underscoring the critical need for advanced research to uncover new therapeutic approaches. Stress granules (SGs) that is implicated in cellular self-protection mechanism, along with its associated family molecules have shown pro-cancer effects and are closely related to tumor chemotherapy resistance. In this study we investigated the relationship between Ras GTPase-activating protein-binding proteins 2 (G3BP2), a core component of SGs, and the malignancy of PDAC as well as its resistance to the chemotherapy drug gemcitabine. Analyzing TCGA dataset revealed that the expression of G3BP1 and G3BP2 was significantly upregulated in PDAC compared with adjacent normal pancreatic tissues, and the high expression of G3BP2 rather than G3BP1 was significantly associated with poorer overall survival (OS) in PDAC patients. We demonstrated that knockdown of G3BP2 inhibited the proliferation and invasion of PANC-1 and CFPAC-1 cells in vitro and in vivo. By analyzing the differentially expressed genes in G3BP2 knockdown and overexpressed PANC-1 cells, we identified DKC1 that was associated with RNA stability and regulation as the target of G3BP2. We demonstrated that G3BP2 bound to PDIA3 mRNA and recruited them into SGs, increasing the stability of PDIA3 mRNA and attenuating its translation efficiency, thereby promoting DKC1 expression. Furthermore, DKC1 could bind to hENT mRNA and inhibited its expression, which enhanced gemcitabine resistance of PDAC. Therefore, we propose a novel mechanism wherein G3BP2 facilitates PDAC's resistance to chemotherapy by modulating PDIA3-DKC1-hENT in a SGs-dependent way, suggesting G3BP2 SGs a protentional therapeutic target for the treatment in PDAC.
Collapse
MESH Headings
- Gemcitabine
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Deoxycytidine/therapeutic use
- Humans
- Drug Resistance, Neoplasm
- Protein Disulfide-Isomerases/metabolism
- Protein Disulfide-Isomerases/genetics
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/genetics
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/genetics
- Cell Line, Tumor
- Animals
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Stress Granules/metabolism
- Cell Cycle Proteins/metabolism
- Cell Cycle Proteins/genetics
- Mice, Nude
- Cell Proliferation/drug effects
- RNA Recognition Motif Proteins/metabolism
- Mice
- Antimetabolites, Antineoplastic/pharmacology
- Antimetabolites, Antineoplastic/therapeutic use
- RNA Helicases/metabolism
- RNA Helicases/genetics
- Poly-ADP-Ribose Binding Proteins/metabolism
- Poly-ADP-Ribose Binding Proteins/genetics
- Mice, Inbred BALB C
- Female
- RNA-Binding Proteins
Collapse
Affiliation(s)
- Fa-Liang Xing
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Bo-Rui Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Ying-Jin Fang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chen Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xian-Jun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
115
|
Dai S, Peng Y, Wang G, Chen C, Chen Q, Yin L, Yan H, Zhang K, Tu M, Lu Z, Wei J, Li Q, Wu J, Jiang K, Zhu Y, Miao Y. LIM domain only 7: a novel driver of immune evasion through regulatory T cell differentiation and chemotaxis in pancreatic ductal adenocarcinoma. Cell Death Differ 2025; 32:271-290. [PMID: 39143228 PMCID: PMC11803110 DOI: 10.1038/s41418-024-01358-7] [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: 04/02/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024] Open
Abstract
With advancements in genomics and immunology, immunotherapy has emerged as a revolutionary strategy for tumor treatment. However, pancreatic ductal adenocarcinoma (PDAC), an immunologically "cold" tumor, exhibits limited responsiveness to immunotherapy. This study aimed to address the urgent need to uncover PDAC's immune microenvironment heterogeneity and identify the molecular mechanisms driving immune evasion. Using single-cell RNA sequencing datasets and spatial proteomics, we discovered LIM domain only 7 (LMO7) in PDAC cells as a previously unrecognized driver of immune evasion through Treg cell enrichment. LMO7 was positively correlated with infiltrating regulatory T cells (Tregs) and dysfunctional CD8+ T cells. A series of in vitro and in vivo experiments demonstrated LMO7's significant role in promoting Treg cell differentiation and chemotaxis while inhibiting CD8+ T cells and natural killer cell cytotoxicity. Mechanistically, LMO7, through its LIM domain, directly bound and promoted the ubiquitination and degradation of Foxp1. Foxp1 negatively regulated transforming growth factor-beta (TGF-β) and C-C motif chemokine ligand 5 (CCL5) expression by binding to sites 2 and I/III, respectively. Elevated TGF-β and CCL5 levels contribute to Treg cell enrichment, inducing immune evasion in PDAC. Combined treatment with TGF-β/CCL5 antibodies, along with LMO7 inhibition, effectively reversed immune evasion in PDAC, activated the immune response, and prolonged mouse survival. Therefore, this study identified LMO7 as a novel facilitator in driving immune evasion by promoting Treg cell enrichment and inhibiting cytotoxic effector functions. Targeting the LMO7-Foxp1-TGF-β/CCL5 axis holds promise as a therapeutic strategy for PDAC. Graphical abstract revealing LMO7 as a novel facilitator in driving immune evasion by promoting Tregs differentiation and chemotaxis, inducing CD8+ T/natural killer cells inhibition.
Collapse
Affiliation(s)
- Shangnan Dai
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Yunpeng Peng
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Guangfu Wang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Chongfa Chen
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Qiuyang Chen
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Lingdi Yin
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Han Yan
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Kai Zhang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Min Tu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Zipeng Lu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Jishu Wei
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Qiang Li
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Junli Wu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Kuirong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China
| | - Yi Zhu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China.
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China.
| | - Yi Miao
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, PR China.
- Pancreas Institute, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, PR China.
- Pancreas Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
116
|
Delis S, Chrysikos D, Liatsos D, Sperdouli D, Alifieris K, Shihada A, Saintanis M, Kokoroskos N, Palantzas A, Liakopoulos K, Troupis T. Warshaw Technique in Spleen-Preserving Pancreatectomy: A Case Series. Cureus 2025; 17:e79211. [PMID: 40115673 PMCID: PMC11924290 DOI: 10.7759/cureus.79211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2025] [Indexed: 03/23/2025] Open
Abstract
We present three cases of spleen-preserving total pancreatectomy using the Warshaw technique for neoplasms of the pancreatic head. The first case involved a 5 cm mucinous pancreatic neoplasm invading the middle colic artery. The second case featured a patient with a BMI of 40 and adenocarcinoma of the pancreatic head. The third case involved a serous cystadenoma in the pancreatic head in a patient with a history of gastric surgery for a gastric ulcer, requiring surgical intervention due to weight loss and abdominal symptoms. These cases highlight the surgical nuances and benefits of the Warshaw technique, particularly its role in preserving splenic blood supply and reducing postoperative complications.
Collapse
Affiliation(s)
- Spiros Delis
- Department of General Surgery, Konstantopoulio General Hospital, Athens, GRC
| | - Dimosthenis Chrysikos
- Department of Anatomy, National and Kapodistrian University of Athens School of Medicine, Athens, GRC
| | - Dimitris Liatsos
- Department of Anatomy, National and Kapodistrian University of Athens School of Medicine, Athens, GRC
| | - Despoina Sperdouli
- Department of General Surgery, Konstantopoulio General Hospital, Athens, GRC
| | | | - Amir Shihada
- Department of Anatomy, National and Kapodistrian University of Athens School of Medicine, Athens, GRC
| | - Michail Saintanis
- Department of Anatomy, National and Kapodistrian University of Athens School of Medicine, Athens, GRC
| | - Nikos Kokoroskos
- Department of General Surgery, Konstantopoulio General Hospital, Athens, GRC
| | - Andreas Palantzas
- Department of Anatomy, National and Kapodistrian University of Athens School of Medicine, Athens, GRC
| | - Konstantinos Liakopoulos
- Department of Anatomy, National and Kapodistrian University of Athens School of Medicine, Athens, GRC
| | - Theodore Troupis
- Department of Anatomy, National and Kapodistrian University of Athens School of Medicine, Athens, GRC
| |
Collapse
|
117
|
Qin J, Liu J, Wei Z, Li X, Chen Z, Li J, Zheng W, Liu H, Xu S, Yong T, Zhao B, Gou S, Ju S, Teng GJ, Yang X, Gan L. Targeted intervention in nerve-cancer crosstalk enhances pancreatic cancer chemotherapy. NATURE NANOTECHNOLOGY 2025; 20:311-324. [PMID: 39496914 DOI: 10.1038/s41565-024-01803-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 09/04/2024] [Indexed: 11/06/2024]
Abstract
Nerve-cancer crosstalk has gained substantial attention owing to its impact on tumour growth, metastasis and therapy resistance. Effective therapeutic strategies targeting tumour-associated nerves within the intricate tumour microenvironment remain a major challenge in pancreatic cancer. Here we develop Escherichia coli Nissle 1917-derived outer membrane vesicles conjugated with nerve-binding peptide NP41, loaded with the tropomyosin receptor kinase (Trk) inhibitor larotrectinib (Lar@NP-OMVs) for tumour-associated nerve targeting. Lar@NP-OMVs achieve efficient nerve intervention to diminish neurite growth by disrupting the neurotrophin/Trk signalling pathway. Moreover, OMV-mediated repolarization of M2-like tumour-associated macrophages to an M1-like phenotype results in nerve injury, further accentuating Lar@NP-OMV-induced nerve intervention to inhibit nerve-triggered proliferation and migration of pancreatic cancer cells and angiogenesis. Leveraging this strategy, Lar@NP-OMVs significantly reduce nerve infiltration and neurite growth promoted by gemcitabine within the tumour microenvironment, leading to augmented chemotherapy efficacy in pancreatic cancer. This study sheds light on a potential avenue for nerve-targeted therapeutic intervention for enhancing pancreatic cancer therapy.
Collapse
Affiliation(s)
- Jiaqi Qin
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjie Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaohan Wei
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaoxia Chen
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jianye Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Wenxia Zheng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Haojie Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Shiyi Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, China
| | - Ben Zhao
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Shanmiao Gou
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shenghong Ju
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Gao-Jun Teng
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China.
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, China.
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
118
|
Moustakis C, Blanck O, Grohmann M, Albers D, Bartels D, Bathen B, Borzì GR, Broggi S, Bruschi A, Casale M, Delana A, Doolan P, Ebrahimi Tazehmahalleh F, Fabiani S, Falco MD, Fehr R, Friedlein M, Gutser S, Hamada AM, Hancock T, Köhn J, Kornhuber C, Krieger T, Lambrecht U, Lappi S, Moretti E, Mirus A, Muedder T, Plaude S, Polvika B, Ravaglia V, Righetto R, Rinaldin G, Schachner H, Scaggion A, Schilling P, Szeverinski P, Villaggi E, Walke M, Wilke L, Winkler P, Nicolay NH, Eich HT, Gkika E, Brunner TB, Schmitt D. Planning Benchmark Study for Stereotactic Body Radiation Therapy of Pancreas Carcinomas With Simultaneously Integrated Boost and Protection: Results of the DEGRO/DGMP Working Group on Stereotactic Radiation Therapy and Radiosurgery. Int J Radiat Oncol Biol Phys 2025; 121:547-557. [PMID: 39222825 DOI: 10.1016/j.ijrobp.2024.08.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/14/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE The proximity or overlap of planning target volume (PTV) and organs-at-risk (OARs) poses a major challenge in stereotactic body radiation therapy (SBRT) of pancreatic cancer (PACA). This international treatment planning benchmark study investigates whether simultaneous integrated boost (SIB) and simultaneous integrated protection (SIP) concepts in PACA SBRT can lead to improved and harmonized plan quality. METHODS AND MATERIALS A multiparametric specification of desired target doses (gross target volume [GTV]D50%, GTVD99%, PTVD95%, and PTV0.5cc) with 2 prescription doses of GTVD50% = 5 × 9.2Gy (46 Gy) and GTVD50% = 8 × 8.25 Gy (66 Gy) and OAR limits were distributed with planning computed tomography and contours from 3 PACA patients. In phase 1, plans were ranked using a scoring system for comparison of trade-offs between GTV/PTV and OAR. In phase 2, replanning was performed for the most challenging case and prescription with dedicated SIB and SIP contours provided for optimization after group discussion. RESULTS For all 3 cases and both phases combined, 292 plans were generated from 42 institutions in 5 countries using commonly available treatment planning systems. The GTVD50% prescription was performed by only 76% and 74% of planners within 2% for 5 and 8 fractions, respectively. The GTVD99% goal was mostly reached, while the balance between OAR and target dose showed initial SIB/SIP-like optimization strategies in about 50% of plans. For plan ranking, 149 and 217 score penalties were given for 5 and 8 fractions, pointing to improvement possibilities. For phase 2, the GTVD50% prescription was performed by 95% of planners within 2%, and GTVD99% as well as OAR doses were better harmonized with notable less score penalties. Fourteen of 19 planners improved their plan rank, 9 of them by at least 2 ranks. CONCLUSIONS Dedicated SIB/SIP concepts in combination with multiparametric prescriptions and constraints can lead to overall harmonized and high treatment plan quality for PACA SBRT. Standardized SIB/SIP treatment planning in multicenter clinical trials appears feasible after group consensus and training.
Collapse
Affiliation(s)
- Christos Moustakis
- University Hospital Leipzig, Department of Radiation Oncology, Leipzig, Germany; University Hospital Muenster, Department of Radiation Oncology, Muenster, Germany.
| | - Oliver Blanck
- University Medical Center Schleswig Holstein, Kiel, Department of Radiation Oncology, Kiel, Germany; Saphir Radiosurgery Center, Frankfurt and Kiel, Germany
| | - Maximilian Grohmann
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dirk Albers
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dennis Bartels
- Department of Radiation Oncology Harburg, Hamburg, Germany
| | - Bastian Bathen
- Department of Radiation Oncology, University Hospital, Goethe University, Frankfurt, Germany; Saphir Radiosurgery Center, Frankfurt, Germany
| | | | | | | | | | - Anna Delana
- Medical Physics Unit, "S. Chiara" Hospital, Trento, Italy
| | | | - Fatemeh Ebrahimi Tazehmahalleh
- Helios Hospital Schwerin, Department of Radiation Oncology, Schwerin, Germany; University Hospital Cologne, Department of Radiation Oncology, Cologne, Germany
| | | | - Maria Daniela Falco
- Department of Radiation Oncology, "G.D'Annunzio" University, "SS.Annunziata" Hospital, Chieti, Italy
| | - Roman Fehr
- University Medicine Rostock, Department of Radiation Oncology, Rostock, Germany
| | - Melissa Friedlein
- Department of Radiation Oncology, University Hospital Jena, Jena, Germany
| | - Susanne Gutser
- Department of Radiation Oncology, University Hospital Augsburg, Augsburg, Germany
| | - Abdul Malek Hamada
- University Medicine Rostock, Department of Radiation Oncology, Rostock, Germany
| | | | - Janett Köhn
- Department of Radiation Oncology, University Hospital, Goethe University, Frankfurt, Germany; Saphir Radiosurgery Center, Frankfurt, Germany
| | - Christine Kornhuber
- University Hospital Halle, Department of Radiation Oncology, Halle (Saale), Germany
| | - Thomas Krieger
- Department of Radiation Oncology, University Hospital Würzburg, Würzburg, Germany
| | - Ulrike Lambrecht
- Department of Radiation Oncology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | | | | | - Thomas Muedder
- Department of Radiation Oncology, University Hospital Bonn, Bonn, Germany
| | - Sandija Plaude
- West German Proton Therapy Center Essen (WPE), Essen, Germany
| | | | | | - Roberto Righetto
- Proton Therapy Unit, S. Chiara Hospital - Azienda Provinciale per I Servizi Sanitari (APSS), Trento, Italy
| | | | - Henrik Schachner
- Department of Radiation Oncology at Weilheim Hospital, Weilheim, Germany
| | | | - Philipp Schilling
- Municipal Hospital Dresden-Friedrichstadt - MVZ Radiotherapy, Dresden, Germany
| | - Philipp Szeverinski
- Institute of Medical Physics, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | | | - Mathias Walke
- University Hospital Magdeburg, Department of Radiation Oncology, Magdeburg, Germany
| | - Lotte Wilke
- University Hospital Zürich, Department of Radiation Oncology, Zürich, Switzerland
| | - Peter Winkler
- University Medical Center Graz, Department of Radiation Oncology, Graz, Austria
| | - Nils H Nicolay
- University Hospital Leipzig, Department of Radiation Oncology, Leipzig, Germany; University Hospital of Freiburg, Department of Radiation Oncology, Freiburg, Germany
| | - Hans Theodor Eich
- University Hospital Muenster, Department of Radiation Oncology, Muenster, Germany
| | - Eleni Gkika
- Department of Radiation Oncology, University Hospital Bonn, Bonn, Germany; University Hospital of Freiburg, Department of Radiation Oncology, Freiburg, Germany
| | - Thomas B Brunner
- University Hospital Magdeburg, Department of Radiation Oncology, Magdeburg, Germany; University Medical Center Graz, Department of Radiation Oncology, Graz, Austria
| | - Daniela Schmitt
- University Medical Center Göttingen, Department of Radiation Oncology, Göttingen, Germany
| |
Collapse
|
119
|
Safari MH, Rahimzadeh P, Alaei E, Alimohammadi M, Esfandiari N, Daneshi S, Malgard N, Farahani N, Taheriazam A, Hashemi M. Targeting ferroptosis in gastrointestinal tumors: Interplay of iron-dependent cell death and autophagy. Mol Cell Probes 2025; 79:102013. [PMID: 39837469 DOI: 10.1016/j.mcp.2025.102013] [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: 11/22/2024] [Revised: 01/06/2025] [Accepted: 01/18/2025] [Indexed: 01/23/2025]
Abstract
Ferroptosis is a regulated cell death mechanism distinct from apoptosis, autophagy, and necroptosis, marked by iron accumulation and lipid peroxidation. Since its identification in 2012, it has developed into a potential therapeutic target, especially concerning GI disorders like PC, HCC, GC, and CRC. This interest arises from the distinctive role of ferroptosis in the progression of diseases, presenting a new avenue for treatment where existing therapies fall short. Recent studies emphasize the promise of focusing on ferroptosis to fight GI cancers, showcasing its unique pathophysiological mechanisms compared to other types of cell death. By comprehending how ferroptosis aids in the onset and advancement of GI diseases, scientists aim to discover novel drug targets and treatment approaches. Investigating ferroptosis in gastrointestinal disorders reveals exciting possibilities for novel therapies, potentially revolutionizing cancer treatment and providing renewed hope for individuals affected by these tumors.
Collapse
Affiliation(s)
- Mohamad Hosein Safari
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elmira Alaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Negin Esfandiari
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Neda Malgard
- Department of Internal Medicine, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
120
|
Zhou T, Niu Y, Li Y. Advances in research on malignant tumors and targeted agents for TOP2A (Review). Mol Med Rep 2025; 31:50. [PMID: 39670307 DOI: 10.3892/mmr.2024.13415] [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: 07/18/2024] [Accepted: 11/28/2024] [Indexed: 12/14/2024] Open
Abstract
The DNA topoisomerase isoform topoisomerase IIα (TOP2A) is essential for the condensation and segregation of cellular mitotic chromosomes and the structural maintenance. It has been demonstrated that TOP2A is highly expressed in various malignancies, including lung adenocarcinoma (LUAD), hepatocellular carcinoma (HCC) and breast cancer (BC), associating with poor prognosis and aggressive tumor behavior. Additionally, TOP2A has emerged as a promising target for cancer therapy, with widespread clinical application of associated chemotherapeutic agents. The present study explored the impact of TOP2A on malignant tumor growth and the advancements in research on its targeted drugs. The fundamental mechanisms of TOP2A have been detailed, its specific roles in tumor cells are analyzed, and its potential as a biomarker for tumor prognosis and therapeutic targeting is highlighted. Additionally, the present review compiles findings from the latest clinical trials of relevant targeted agents, information on newly developed inhibitors, and discusses future research directions and clinical application strategies in cancer therapy, aiming to propose novel ideas and methods.
Collapse
Affiliation(s)
- Tao Zhou
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| | - Yiting Niu
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| | - Yanjun Li
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| |
Collapse
|
121
|
Baghel K, Mehrotra S, Prajapati VK. Revolutionizing pancreatic cancer treatment with CAR-T therapy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:331-353. [PMID: 39978971 DOI: 10.1016/bs.apcsb.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Pancreatic cancer remains one of the most lethal malignancies, with a five-year survival rate among the lowest of all cancers. This poor prognosis is largely due to the aggressive nature of the disease and its resistance to conventional treatments such as surgery, chemotherapy, and radiation therapy. Chimeric antigen receptor (CAR) T-cell therapy, a novel immunotherapeutic approach leverages the patient's own immune system to specifically target and eliminate cancer cells by genetically engineering T cells to express CARs that recognize tumor-specific antigens. While CAR-T therapy has demonstrated remarkable success in treating hematologic malignancies, its application to solid tumors like pancreatic cancer presents significant challenges. Recent advancements in CAR-T cell design, like the addition of co-stimulatory domains and dual-targeting CARs, have enhanced their efficacy against solid tumors. Additionally, strategies to modify the tumor microenvironment (TME), such as combining CAR-T therapy with immune checkpoint inhibitors and cytokine modulation, are being investigated to boost CAR-T cell activity against pancreatic cancer. Early-phase clinical trials targeting antigens such as carcinoembryonic antigen (CEA) and mesothelin (MSLN) in pancreatic cancer have yielded encouraging results, though obstacles like antigen escape and limited T-cell persistence remain significant challenges. This chapter outlines the current state of CAR-T therapy for pancreatic cancer, focusing on the emerging approaches to address these obstacles and underscore the potential of CAR-T therapy to transform the future of pancreatic cancer treatment.
Collapse
Affiliation(s)
- Kirti Baghel
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
122
|
Tan Z, Meng Y, Wu Y, Zhen J, He H, Pu Y, Zhang J, Dong W. The burden and temporal trend of early onset pancreatic cancer based on the GBD 2021. NPJ Precis Oncol 2025; 9:32. [PMID: 39880919 PMCID: PMC11779834 DOI: 10.1038/s41698-025-00820-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
In the context of the global increase in early-onset tumours, investigating the global disease burden caused by early-onset pancreatic cancer (EOPC) is imperative. Data on the burden of EOPC were obtained from the Global Burden of Disease Study 2021. A joinpoint regression model was used to analyse the temporal trend of the EOPC burden, and an age‒period‒cohort (APC) model was used to analyse the influence of age, period, and birth cohort on burden trends. Globally, the number of EOPC cases increased from 24,480 to 42,254, and the number of deaths increased from 17,193 to 26,996 between 1990 and 2021. The results of the APC model showed that the burden of EOPC increases with increasing age, whereas the variations in period and cohort effects exhibited a complex pattern across different sociodemographic index regions. Consequently, the disease burden of EOPC is increasing worldwide, highlighting the need for effective interventions.
Collapse
Affiliation(s)
- Zongbiao Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Yang Meng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yanrui Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Junhai Zhen
- Department of General Practice, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, China
| | - Haodong He
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Yu Pu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Jixiang Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
| |
Collapse
|
123
|
He H, Chen G, Tang Z, Chen CYC. Dual modality feature fused neural network integrating binding site information for drug target affinity prediction. NPJ Digit Med 2025; 8:67. [PMID: 39875637 PMCID: PMC11775287 DOI: 10.1038/s41746-025-01464-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025] Open
Abstract
Accurately predicting binding affinities between drugs and targets is crucial for drug discovery but remains challenging due to the complexity of modeling interactions between small drug and large targets. This study proposes DMFF-DTA, a dual-modality neural network model integrates sequence and graph structure information from drugs and proteins for drug-target affinity prediction. The model introduces a binding site-focused graph construction approach to extract binding information, enabling more balanced and efficient modeling of drug-target interactions. Comprehensive experiments demonstrate DMFF-DTA outperforms state-of-the-art methods with significant improvements. The model exhibits excellent generalization capabilities on completely unseen drugs and targets, achieving an improvement of over 8% compared to existing methods. Model interpretability analysis validates the biological relevance of the model. A case study in pancreatic cancer drug repurposing demonstrates its practical utility. This work provides an interpretable, robust approach to integrate multi-view drug and protein features for advancing computational drug discovery.
Collapse
Affiliation(s)
- Haohuai He
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Sun Yat-sen University, Shenzhen, 510275, China
| | - Guanxing Chen
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Sun Yat-sen University, Shenzhen, 510275, China
| | - Zhenchao Tang
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Sun Yat-sen University, Shenzhen, 510275, China
| | - Calvin Yu-Chian Chen
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
- Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan.
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 41354, Taiwan.
| |
Collapse
|
124
|
Chen P, Hou W, Li C, Liang Q, Ma L, Zhao X, Yi C. Lived experiences of patients with advanced pancreatic cancer on patient-reported outcomes (PROs) management: a qualitative phenomenological study in Southwest China. BMJ Open 2025; 15:e084259. [PMID: 39880447 PMCID: PMC11781101 DOI: 10.1136/bmjopen-2024-084259] [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: 01/14/2024] [Accepted: 01/08/2025] [Indexed: 01/31/2025] Open
Abstract
OBJECTIVES To explore the lived experiences of patients with advanced pancreatic cancer enrolled in a patient-reported outcomes (PROs) management programme and to preliminarily understand how PROs management influences various aspects of patient care and overall quality of life. DESIGN A qualitative phenomenological study. SETTING A national cancer care centre in Southwest China specialised in cancer care, with a comprehensive PROs management programme. PARTICIPANTS 15 participants diagnosed with advanced pancreatic cancer. RESULTS Five key themes emerged from our interviews, including enhanced communication with healthcare providers, attributed to the structured nature of PROs; increased perceived sense of care, with patients feeling more valued and heard; PROs management facilitated better treatment decision-making, with patients feeling more involved and empowered; improved communication with family members, aiding in better understanding and support; and varied perceptions of the impact on quality of life, with some noting improvements in specific aspects like symptom management, while others were uncertain about the overall benefit. CONCLUSIONS PROs management plays a significant role in improving communication between patients with advanced pancreatic cancer and their healthcare providers, enhancing patients' involvement in treatment decisions, and potentially improving family dynamics. However, the impact of PROs management on the overall quality of life of patients remains complex and individualised. The findings suggest that further research with a more diverse patient population is needed to fully understand the implications of PROs management in advanced cancer care.
Collapse
Affiliation(s)
- Ping Chen
- Department of Oncology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan, China
| | - Wanting Hou
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Changlin Li
- Department of Oncology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan, China
| | - Qingyue Liang
- Clinical Nutrition Department, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan, China
| | - Li Ma
- Department of Oncology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan, China
| | - Xiumei Zhao
- Department of Oncology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, Sichuan, China
| | - Cheng Yi
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
125
|
Rompianesi G, Loiaco G, Rescigno L, Benassai G, Giglio MC, Campanile S, Caggiano M, Montalti R, Troisi RI. A Systematic Review of Indications and Clinical Outcomes of Electrochemotherapy in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2025; 17:408. [PMID: 39941777 PMCID: PMC11816056 DOI: 10.3390/cancers17030408] [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: 12/24/2024] [Revised: 01/21/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most difficult cancers to treat, with a dismal 5-year survival rate of only 8-10%. This challenging prognosis highlights the urgent need for innovative therapeutic approaches to improve outcomes for patients with PDAC. Electrochemotherapy (ECT), which enhances intracellular chemotherapeutic uptake via electric pulses, has been explored for resectable, borderline resectable (BR), locally advanced (LA), recurrent, and metastatic PDAC, either as a complement to conventional treatments or as an alternative when these are not feasible or effective, offering possible benefits in symptomatic palliation and local tumor control. METHODS A systematic review was performed in accordance with PRISMA guidelines for studies assessing the efficacy of ECT in PDAC. After searching Embase, PubMed/MEDLINE, Scopus, and Web of Science, five studies with a combined total of 43 patients in various disease stages were identified. RESULTS ECT showed promise in improving tumor control, alleviating cancer-related pain, and improving quality of life. One study noted a trend towards tumor size reduction of 8.3% at one-month and 16.1% at six-months follow-up (p = 0.211 and p = 0.315), although these findings were derived from studies conducted without specific comparative control groups. Severity of complication was mainly mild (Clavien-Dindo I-II), while severe complications occurred in only 2.3% of patients. Median overall survival was reported in two studies as 8 months (range 2-19) and 11.5 months (range 1-74). ECT showed efficacy for symptom management, with 60% of patients reporting reduced pain/discomfort and 40% showing enhanced quality of life in one study, while another reported pain scores as decreasing from 6 to 3 at one month and to 2 at six months. CONCLUSIONS ECT appears to be a new promising and safe adjunct treatment modality in PDAC management across different disease stages, with potential benefits in tumor control, cancer-related pain reduction, and quality of life. Further studies are warranted to validate these findings and identify patients who could benefit most.
Collapse
Affiliation(s)
- Gianluca Rompianesi
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive and Robotic HPB S and Gery, Transplantation Service, Federico II University Hospital, Via Sergio Pansini 5, 80131 Naples, Italy; (G.R.); (G.L.); (L.R.); (G.B.); (M.C.G.); (S.C.); (M.C.); (R.M.)
| | - Giuseppe Loiaco
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive and Robotic HPB S and Gery, Transplantation Service, Federico II University Hospital, Via Sergio Pansini 5, 80131 Naples, Italy; (G.R.); (G.L.); (L.R.); (G.B.); (M.C.G.); (S.C.); (M.C.); (R.M.)
| | - Luigi Rescigno
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive and Robotic HPB S and Gery, Transplantation Service, Federico II University Hospital, Via Sergio Pansini 5, 80131 Naples, Italy; (G.R.); (G.L.); (L.R.); (G.B.); (M.C.G.); (S.C.); (M.C.); (R.M.)
| | - Gianluca Benassai
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive and Robotic HPB S and Gery, Transplantation Service, Federico II University Hospital, Via Sergio Pansini 5, 80131 Naples, Italy; (G.R.); (G.L.); (L.R.); (G.B.); (M.C.G.); (S.C.); (M.C.); (R.M.)
| | - Mariano Cesare Giglio
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive and Robotic HPB S and Gery, Transplantation Service, Federico II University Hospital, Via Sergio Pansini 5, 80131 Naples, Italy; (G.R.); (G.L.); (L.R.); (G.B.); (M.C.G.); (S.C.); (M.C.); (R.M.)
| | - Silvia Campanile
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive and Robotic HPB S and Gery, Transplantation Service, Federico II University Hospital, Via Sergio Pansini 5, 80131 Naples, Italy; (G.R.); (G.L.); (L.R.); (G.B.); (M.C.G.); (S.C.); (M.C.); (R.M.)
| | - Marcello Caggiano
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive and Robotic HPB S and Gery, Transplantation Service, Federico II University Hospital, Via Sergio Pansini 5, 80131 Naples, Italy; (G.R.); (G.L.); (L.R.); (G.B.); (M.C.G.); (S.C.); (M.C.); (R.M.)
| | - Roberto Montalti
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive and Robotic HPB S and Gery, Transplantation Service, Federico II University Hospital, Via Sergio Pansini 5, 80131 Naples, Italy; (G.R.); (G.L.); (L.R.); (G.B.); (M.C.G.); (S.C.); (M.C.); (R.M.)
- Department of Public Health, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Roberto Ivan Troisi
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive and Robotic HPB S and Gery, Transplantation Service, Federico II University Hospital, Via Sergio Pansini 5, 80131 Naples, Italy; (G.R.); (G.L.); (L.R.); (G.B.); (M.C.G.); (S.C.); (M.C.); (R.M.)
| |
Collapse
|
126
|
Yu L, Guo Q, Li Y, Mao M, Liu Z, Li T, Wang L, Zhang X. CHMP4C promotes pancreatic cancer progression by inhibiting necroptosis via the RIPK1/RIPK3/MLKL pathway. J Adv Res 2025:S2090-1232(25)00058-X. [PMID: 39870301 DOI: 10.1016/j.jare.2025.01.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/10/2025] [Accepted: 01/24/2025] [Indexed: 01/29/2025] Open
Abstract
INTRODUCTION Pancreatic cancer (PC) cannot currently be completely cured and has a poor prognosis. Necroptosis is a distinct form of regulated cell death that differs from both necrosis and apoptosis. Understanding the role of necroptosis during PC progression would open new avenues for targeted therapy. OBJECTIVES The purpose of this study is to examine the impact of necroptosis on the progression of PC and related mechanisms. METHODS RNA sequencing was performed to identify necroptosis-related genes that are differentially expressed in PC tissues. The biological functions of CHMP4C and its necroptosis effects were determined in vitro and in vivo. RNA immunoprecipitation, MeRIP-qPCR, Co-immunoprecipitation assays were conducted to evaluate the interaction among CHMP4C, YBX1 and caspase-8 mRNA. Extracellular vesicles were isolated using the differential ultracentrifugation method. The expression of CHMP4C, p-MLKL and CD117 were detected on a PC tissue microarray using multiplex immunofluorescence staining. RESULTS CHMP4C was significantly overexpressed in PC cells and tissues. It promoted cell growth and suppressed necroptosis of PC cells in both in vivo and in vitro settings. Mechanistically, CHMP4C interacted with YBX1 to mediate m5C modification of caspase-8 mRNA, resulting in increased caspase-8 expression and inhibition of RIPK1/RIPK3/MLKL pathway phosphorylation. Furthermore, CHMP4C promoted extracellular exocytosis of p-MLKL to further suppress necroptosis. Additionally, PC cells used CHMP4C within extracellular vesicles to recruit and stimulate mast cells (MCs), which in turn promoted PC cell proliferation. In PC tissues, the expression of CHMP4C showed a negative correlation with p-MLKL and a positive association with CD117. High expression levels of CHMP4C in patients were associated with poorer overall survival outcomes. CONCLUSIONS CHMP4C promotes PC progression by inhibiting necroptosis, which has potential as a biomarker and therapeutic target in PC.
Collapse
Affiliation(s)
- Longchen Yu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012 China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012 China
| | - Qining Guo
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012 China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012 China
| | - Yaping Li
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012 China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012 China
| | - Mai Mao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012 China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012 China
| | - Zhenping Liu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012 China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012 China
| | - Tingting Li
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012 China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012 China
| | - Lei Wang
- Department of Orthodontics, Qilu Hospital of Shandong University, Jinan 250012 China.
| | - Xin Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012 China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012 China.
| |
Collapse
|
127
|
Xiao X, Huang P, Xu XT. The choice of adjuvant radiotherapy in pancreatic cancer patients after up-front radical surgery. PLoS One 2025; 20:e0317995. [PMID: 39854493 PMCID: PMC11760613 DOI: 10.1371/journal.pone.0317995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND The role of adjuvant radiotherapy in pancreatic cancer following radical surgery remains a subject of of controversy. This study aimed to more accurately screen pancreatic patients who benefit from adjuvant radiotherapy. METHODS Clinicopathologic characteristics of patients with resectable pancreatic cancer were collected from the Surveillance, Epidemiology, and End Results (SEER) database (2004-2015). Univariate and multivariate analyses were applied to identify prognostic factors affecting patient survival. All the patients were divided into two groups, one receiving radiation and the other not. Selection bias were reduced by propensity-score matching (PSM). Kaplan-Meier analysis was used to estimate overall survival (OS) and cancer-specific survival (CSS) between the two groups. RESULTS Within 7097 patients, 2276 received adjuvant radiotherapy (external beam radiation), and 4821 did not. Multivariate analysis revealed that race, age, median income, sex, year of diagnosis, American Joint Committee on Cancer (AJCC) T stage, N stage, scope region lymph surgery, chemotherapy, and radiotherapy were independent predictors for overall survival of all the patients (all p < 0.05). After PSM, a total of 4304 patients were included. There was no OS and CSS benefit of radiotherapy compared with no-radiotherapy (all p > 0.05). Among patients with N1 stage, the radiotherapy group exhibited a median overall survival (mOS) of 21 months (95% CI, 19.82 to 22.18), while the non-radiotherapy group showed a slightly lower mOS of 18 months (95% CI, 16.88 to 19.12). Similarly, in terms of median cancer-specific survival (mCSS), the radiotherapy group demonstrated a mCSS of 22 months (95% CI, 20.79 to 23.21), whereas the non-radiotherapy group had a slightly shorter mCSS of 19 months (95% CI, 17.81 to 20.19). Radiotherapy reduced the all-cause mortality rate and cancer-specific mortality rate among patients with the N1 stage and T4 stage (all p < 0.05). In contrast, the patients in the radiotherapy group with the N0 stage (mOS, 28 months versus 34 months; mCSS, 30 months versus 41months), or primary focus on the body and tail of the pancreas (mOS, 23 months versus 29 months; mCSS, 25 months versus 32 months), or T1 stage (mOS, 36 months versus 113 months; mCSS, 36 months versus 104 months) exhibited a higher all-cause mortality rate and cancer-specific mortality rate compared to those without radiotherapy (all p < 0.05). Subgroup analysis indicated N1 stage pancreatic cancer patients with T2-4 stage, primary focus on the head of the pancreas, young age of onset, and combination chemotherapy were in favor of the adjuvant radiotherapy group (all p < 0.05). CONCLUSIONS Our analysis demonstrates that adjuvant radiotherapy may be beneficial for N1 stage (N+) pancreatic cancer patients who have undergone up-front radical surgery with T2-4 stage, primary focus on the head of the pancreas, young age of onset, and receiving combination chemotherapy. However, radiotherapy needs to be used with caution in patients with T1 stage, N0 stage (N-), or primary focus on the body and tail of the pancreas. These findings may contribute to the development of personalized selection criteria for adjuvant radiotherapy in post-surgical pancreatic cancer patients.
Collapse
Affiliation(s)
- Xia Xiao
- Department of Oncology, Wuxi No.2 People’ s Hospital, Jiangnan University Medical Center, Wuxi, Jiangsu Province, China
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Pei Huang
- Department of Oncology, Wuxi No.2 People’ s Hospital, Jiangnan University Medical Center, Wuxi, Jiangsu Province, China
| | - Xiao-Ting Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
128
|
Chen J, Ma Z, Xu Y, Ge J, Yao H, Li C, Hu X, Pu Y, Li M, Jiang C. CT-based machine learning radiomics predicts Ki-67 expression level and its relationship with overall survival in resectable pancreatic ductal adenocarcinoma. Abdom Radiol (NY) 2025:10.1007/s00261-025-04798-y. [PMID: 39841230 DOI: 10.1007/s00261-025-04798-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND The prognostic prediction of pancreatic ductal adenocarcinoma (PDAC) remains challenging. This study aimed to develop a radiomics model to predict Ki-67 expression status in PDAC patients using radiomics features from dual-phase enhanced CT, and integrated clinical characteristics to create a radiomics-clinical nomogram for prognostic prediction. METHODS In this retrospective study, data were collected from 124 PDAC patients treated surgically at a single center, from January 2017 to March 2023. Patients were categorized according to the Ki-67 expression rate. Radiomics features were extracted from arterial and portal venous phase CT images using 3D Slicer v5.0.3. A radiomics model was formulated and validated to predict the Ki-67 expression, and a nomogram combining clinical indicators and the radiomics model was developed to predict 1, 2 and 3 year overall survival (OS). RESULTS The optimal Ki-67 expression rate cutoff was identified as 50%, with significant OS differences. The developed radiomics model showed good predictive ability with area under the curves of 0.806 and 0.801 in the training and validation groups, respectively. High radiomics score, elevated carbohydrate antigen 19-9 (CA19-9), and receipt of adjuvant chemotherapy were identified as independent prognostic factors for OS. The radiomics-clinical nomogram accurately predicted 1, 2 and 3 year OS in PDAC patients. CONCLUSIONS The radiomics-clinical nomogram provides a non-invasive and efficient method for predicting Ki-67 expression and overall survival in PDAC patients, which could potentially guide clinical decision-making and improve patient outcomes.
Collapse
Affiliation(s)
- Jiahao Chen
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China
| | - Zhuangxuan Ma
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yamin Xu
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China
| | - Jieqiong Ge
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China
| | - Hongfei Yao
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China
| | - Chunjing Li
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China
| | - Xiao Hu
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China
| | - Yunlong Pu
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China
| | - Ming Li
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China.
| | - Chongyi Jiang
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China.
| |
Collapse
|
129
|
Yang Y, Zong S, Hua Y. Nomogram for prognosis prediction in metastatic pancreatic cancer patients undergoing intra-arterial infusion chemotherapy: incorporating immune-inflammation scores and coagulation indicators. BMC Cancer 2025; 25:107. [PMID: 39833690 PMCID: PMC11749238 DOI: 10.1186/s12885-025-13523-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Pancreatic cancer is one of the most malignant tumors with an inferior prognosis. This study aims to determine the prognostic significance of immune-inflammatory scores and coagulation indices in patients with metastatic pancreatic cancer(MPC) and develop a predictive nomogram. METHODS This study retrospectively analyzed the clinical data of 384 patients with MPC who underwent intra-arterial infusion chemotherapy (IAIC). Patients were randomly divided into training and validation cohorts. Firstly, the optimal cutoff values for continuous variables were obtained in the training cohort. Then, survival analysis was performed to evaluate the impact of immune-inflammatory scores neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), systemic immune-inflammation index (SII), and coagulation indicators prothrombin time (PT), fibrinogen (FIB), and D-dimer on the overall survival (OS) of patients. Next, univariate analysis was utilized to identify prognostic factors, and a stepwise regression method was employed for variable selection to construct a nomogram based on the Cox proportional hazards model. Additionally, the predictive performance of the nomogram was assessed by the concordance index (C-index), the area under the ROC curve (AUC), and calibration curves. Finally, patients were stratified into risk groups based on the total score of the nomogram. RESULTS The Kaplan-Meier survival curves indicated that immune-inflammatory scores NLR, PLR, SII, and coagulation indicators PT, FIB, and D-dimer were associated with OS. Through Cox regression analysis, a nomogram was ultimately constructed incorporating NLR, PLR, PT, alkaline phosphatase (ALP), carbohydrate antigen 125 (CA125), age, and ablation. The model demonstrated good discriminative ability, with a C-index of 0.722, and the AUC values at 6- and 12-month OS predictions were 0.828 and 0.851 in the training cohort, while in the validation cohort, the corresponding AUC values were 0.754 and 0.791, respectively. The calibration curves showed a good fit, confirming the stability of the model. A cutoff value of 353.3 was identified as optimal for risk stratification, with a statistically significant difference in OS between the high- and low-risk groups. CONCLUSION The nomogram based on immune-inflammatory scores, coagulation indicators, and other clinicopathological factors can effectively predict the OS of patients with MPC.
Collapse
Affiliation(s)
- Yifan Yang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Shaoqi Zong
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Yongqiang Hua
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
130
|
Garay MI, Mazo T, Ferrero V, Barotto NN, Lagares C, Granton MF, Moreira-Espinoza MJ, Cremonezzi DC, Comba A, Brunotto MN, Tolosa EJ, Fernandez-Zapico ME, Pasqualini ME. Novel inhibitory effect of Omega-3 fatty acids regulating pancreatic cancer progression. Carcinogenesis 2025; 46:bgae081. [PMID: 39742417 PMCID: PMC12097994 DOI: 10.1093/carcin/bgae081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 01/03/2025] Open
Abstract
Pancreatic cancer is a devastating malignancy in great need of new and more effective treatment approaches. In recent years, studies have indicated that nutritional interventions, particularly nutraceuticals, may provide novel avenues to modulate cancer progression. Here, our study characterizes the impact of ω-3 polyunsaturated fatty acids, eicosapentaenoic acid, and docosahexaenoic acid, as a nutraceutical intervention in pancreatic cancer using a genetically engineered mouse model driven by KrasG12D and Trp53R172H. This model closely resembles human pancreatic carcinogenesis, offering a disease relevant platform for translational research. Our findings showed that ω-3 polyunsaturated fatty acids intervention (using a diet supplemented with 6% cod liver oil) significantly reduced tumor volume as well as lung and liver metastasis and a trend toward improved survival rate compared with control treated mice. This antitumoral effect was accompanied by distinct changes in tumor membrane fatty acid profile and eicosanoids release. Furthermore, the eicosapentaenoic acid and docosahexaenoic acid intervention also reduced malignant histological parameters and induced apoptosis without affecting cell proliferation. Of note is the significant reduction in tumor fibrosis that was associated with decreased levels of Sonic Hedgehog, a major ligand controlling this cellular compartment in pancreatic cancer. All together our results demonstrate the impact of eicosapentaenoic acid and docosahexaenoic acid as antitumor regulators in pancreatic cancer, suggesting potential for ω-3 polyunsaturated fatty acids as a possible antitumoral dietary intervention. This research opens new avenues for integrating nutraceutical strategies in pancreatic cancer management.
Collapse
Affiliation(s)
- María I Garay
- Instituto de Investigaciones en Ciencias de la Salud, INICSA (CONICET - FCM UNC), 5016 Córdoba, Argentina
- Instituto de Biología Celular y Cátedra de Biología Celular, Histología y Embriología, FCM-UNC, 5016 Córdoba, Argentina
| | - Tamara Mazo
- Instituto de Investigaciones en Ciencias de la Salud, INICSA (CONICET - FCM UNC), 5016 Córdoba, Argentina
- Instituto de Biología Celular y Cátedra de Biología Celular, Histología y Embriología, FCM-UNC, 5016 Córdoba, Argentina
| | - Victoria Ferrero
- Instituto de Investigaciones en Ciencias de la Salud, INICSA (CONICET - FCM UNC), 5016 Córdoba, Argentina
- Instituto de Biología Celular y Cátedra de Biología Celular, Histología y Embriología, FCM-UNC, 5016 Córdoba, Argentina
| | - Nelso N Barotto
- Instituto de Biología Celular y Cátedra de Biología Celular, Histología y Embriología, FCM-UNC, 5016 Córdoba, Argentina
| | - Clarisa Lagares
- Instituto de Investigaciones en Ciencias de la Salud, INICSA (CONICET - FCM UNC), 5016 Córdoba, Argentina
| | - María F Granton
- Instituto de Biología Celular y Cátedra de Biología Celular, Histología y Embriología, FCM-UNC, 5016 Córdoba, Argentina
| | - María J Moreira-Espinoza
- Instituto de Biología Celular y Cátedra de Biología Celular, Histología y Embriología, FCM-UNC, 5016 Córdoba, Argentina
| | - David C Cremonezzi
- Departamento de Patología, Hospital Nacional de Clínicas, FCM-UNC, 5000 Córdoba, Argentina
| | - Andrea Comba
- Department of Pathology, Division Neuropathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, United States
| | - Mabel N Brunotto
- Instituto de Investigaciones en Ciencias de la Salud, INICSA (CONICET - FCM UNC), 5016 Córdoba, Argentina
- Departamento de Biología Bucal, Facultad de Odontología, UNC, 5016 Córdoba, Argentina
| | - Ezequiel J Tolosa
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN 55905, United States
| | | | - María E Pasqualini
- Instituto de Investigaciones en Ciencias de la Salud, INICSA (CONICET - FCM UNC), 5016 Córdoba, Argentina
- Instituto de Biología Celular y Cátedra de Biología Celular, Histología y Embriología, FCM-UNC, 5016 Córdoba, Argentina
| |
Collapse
|
131
|
Zhang RH, Chen T, Xiong QQ, Wang S, Chen GQ, Zhang WL, Yuan HF, Zhao YL, Liu T, Huang Y, Zhou M, Yang CL, Liao SG, Li YJ. Discovery of a potent anticancer agent against pancreatic ductal adenocarcinoma targeting FAK with DFG-out state and JAK/Aurora kinases. Eur J Med Chem 2025; 282:117059. [PMID: 39577230 DOI: 10.1016/j.ejmech.2024.117059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/20/2024] [Accepted: 10/31/2024] [Indexed: 11/24/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a clinically challenging cancer because of the difficulty in diagnosis and its resistance to chemotherapy. Focal adhesion kinase (FAK) is found overexpressed in PDAC, and targeting FAK has been proved to impede the progress of PDAC. However, most of FAK inhibitors were reported to bind with FAK in a DFG-in conformation, leading to a limited anti-tumor effect in clinical studies. Herein, to develop FAK inhibitors targeting the inactive DFG-out conformation, a series of large aromatic rings were selected to improve the interaction with Phe565 of the DFG motif. Compound 26 was designed to effectively inhibit FAK and the proliferation of PANC-1 cells with IC50 of 50.94 nM and 0.15 μM, respectively. Besides, compound 26 was proved to strongly suppress the proliferation, colony formation, migration, and invasion in FAK-overexpressing PDAC cells. This inhibitor was confirmed to induce the apoptosis and G2/M arrest in PANC-1 cells through the suppression of FAK/PI3K/Akt signal pathway. Meanwhile, compound 26 was found to simultaneously inhibit FAK with DFG-out conformation and JAK3/Aurora B (IC50 of 9.99 nM and 0.49 nM, respectively). In vivo, compound 26 effectively inhibited the tumorigenesis and metastasis of PDAC with desirable biosafety. Overall, these results suggested that compound 26 was a promising candidate for the treatment of PDAC.
Collapse
Affiliation(s)
- Rong-Hong Zhang
- Center for Tissue Engineering and Stem Cell Research, Key Laboratory of Regenerative Medicine of Guizhou Province, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, China; State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou, China
| | - Ting Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou, China
| | - Qian-Qian Xiong
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou, China
| | - Shan Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou, China
| | - Guo-Qi Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou, China
| | - Wen-Li Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou, China
| | - Hong-Fei Yuan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou, China
| | - Yong-Long Zhao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou, China
| | - Ting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou, China
| | - Yong Huang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou, China
| | - Meng Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou, China.
| | - Cheng-Li Yang
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| | - Shang-Gao Liao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou, China.
| | - Yong-Jun Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), School of Pharmacy, Guizhou Medical University, Guian New District, Guizhou, China.
| |
Collapse
|
132
|
Oyon D, Lopez-Pascual A, Castello-Uribe B, Uriarte I, Orsi G, Llorente S, Elurbide J, Adan-Villaescusa E, Valbuena-Goiricelaya E, Irigaray-Miramon A, Latasa MU, Martinez-Perez LA, Bonetti LR, Prosper F, Ponz-Sarvise M, Vicent S, Pineda-Lucena A, Ruiz-Clavijo D, Sangro B, Larracoechea UA, Tian TV, Casadei-Gardini A, Amat I, Arechederra M, Berasain C, Urman JM, Avila MA, Fernandez-Barrena MG. Targeting of the G9a, DNMT1 and UHRF1 epigenetic complex as an effective strategy against pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res 2025; 44:13. [PMID: 39810240 PMCID: PMC11734372 DOI: 10.1186/s13046-024-03268-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with limited treatment options and a poor prognosis. The critical role of epigenetic alterations such as changes in DNA methylation, histones modifications, and chromatin remodeling, in pancreatic tumors progression is becoming increasingly recognized. Moreover, in PDAC these aberrant epigenetic mechanisms can also limit therapy efficacy. This study aimed to investigate the expression and prognostic significance of a key epigenetic complex encompassing DNA methyltransferase-1 (DNMT1), the histone methyltransferase G9a, and the scaffold protein UHRF1 in PDAC. We also evaluated the therapeutic potential of an innovative inhibitor targeting these epigenetic effectors. METHODS Immunohistochemical analysis of DNMT1, G9a, and UHRF1 expression was conducted in human PDAC tissue samples. Staining was semi-quantitatively scored, and overexpression was defined as moderate to strong positivity. The prognostic impact was assessed by correlating protein expression with patient survival. The antitumoral effects of the dual DNMT1-G9a inhibitor CM272 were tested in PDAC cell lines, followed by transcriptomic analyses to identify underlying mechanisms. The in vivo antitumoral efficacy of CM272 was evaluated in PDAC xenograft and syngeneic mouse models, both alone and in combination with anti-PD1 immunotherapy. RESULTS DNMT1, G9a, and UHRF1 were significantly overexpressed in PDAC cells and stroma compared to normal pancreatic tissues. Simultaneous overexpression of the three proteins was associated with significantly reduced survival in resected PDAC patients. CM272 exhibited potent antiproliferative activity in PDAC cell lines, inducing apoptosis and altering key metabolic and cell cycle-related genes. CM272 also enhanced chemotherapy sensitivity and significantly inhibited tumor growth in vivo without detectable toxicity. Combination of CM272 with anti-PD1 therapy further improved antitumor responses and immune cell infiltration, particularly CD4 + and CD8 + T cells. CONCLUSIONS The combined overexpression of DNMT1, G9a, and UHRF1 in PDAC is a strong predictor of poor prognosis. CM272, by targeting this epigenetic complex, shows promising therapeutic potential by inducing apoptosis, reprogramming metabolic pathways, and enhancing immune responses. The combination of CM272 with immunotherapy offers a novel, effective treatment strategy for PDAC.
Collapse
Affiliation(s)
- Daniel Oyon
- Department of Gastroenterology, Galdakao-Usansolo Hospital, Galdakao, Spain
- Biobizkaia Health Research Institute, Bizkaia, Spain
| | - Amaya Lopez-Pascual
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
| | - Borja Castello-Uribe
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | - Iker Uriarte
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Giulia Orsi
- Oncology Department, University Hospital of Modena, Modena, Italy
- Department of Oncology, IRCCS San Raffaele Scientific Institute Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Sofia Llorente
- Preclinical and Translational Research Program, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jasmin Elurbide
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Elena Adan-Villaescusa
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | | | - Ainara Irigaray-Miramon
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | - Maria Ujue Latasa
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | - Luz A Martinez-Perez
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- Universidad de Guadalajara Centro Universitario de Ciencias de La Salud, Guadalajara, Mexico
| | - Luca Reggiani Bonetti
- Department of Medical and Surgical Sciences for Children and Adults, Division of Pathology University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Felipe Prosper
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- Hemato-Oncology Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERonc, Instituto de Salud Carlos III, Madrid, Spain
| | - Mariano Ponz-Sarvise
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- CIBERonc, Instituto de Salud Carlos III, Madrid, Spain
- Departments of Oncology and Immunology, Clinica Universidad de Navarra, Pamplona, Spain
| | - Silvestre Vicent
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- CIBERonc, Instituto de Salud Carlos III, Madrid, Spain
- Oncogenes and Effector Targets Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
| | | | - David Ruiz-Clavijo
- Department of Gastroenterology and Hepatology, Navarra University Hospital Complex, Pamplona, Spain
| | - Bruno Sangro
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
- Hepatology Unit, Clínica Universidad de Navarra, CCUN, Pamplona, Spain
| | - Urko Aguirre Larracoechea
- Research Unit, Osakidetza Basque Health Service, Barrualde-Galdakao Integrated Health Organisation, Galdakao-Usansolo Hospital, Galdakao, Spain
| | - Tian V Tian
- Preclinical and Translational Research Program, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Andrea Casadei-Gardini
- Oncology Department, University Hospital of Modena, Modena, Italy
- Department of Oncology, IRCCS San Raffaele Scientific Institute Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Irene Amat
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- Department of Pathology, Navarra University Hospital Complex, Pamplona, Spain
| | - Maria Arechederra
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Berasain
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Jesus M Urman
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- Department of Gastroenterology and Hepatology, Navarra University Hospital Complex, Pamplona, Spain
| | - Matias A Avila
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Maite G Fernandez-Barrena
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain.
- Navarra Institute for Health Research, IdiSNA, Pamplona, Spain.
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
133
|
Zhan T, Betge J, Schulte N, Dreikhausen L, Hirth M, Li M, Weidner P, Leipertz A, Teufel A, Ebert MP. Digestive cancers: mechanisms, therapeutics and management. Signal Transduct Target Ther 2025; 10:24. [PMID: 39809756 PMCID: PMC11733248 DOI: 10.1038/s41392-024-02097-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Cancers of the digestive system are major contributors to global cancer-associated morbidity and mortality, accounting for 35% of annual cases of cancer deaths. The etiologies, molecular features, and therapeutic management of these cancer entities are highly heterogeneous and complex. Over the last decade, genomic and functional studies have provided unprecedented insights into the biology of digestive cancers, identifying genetic drivers of tumor progression and key interaction points of tumor cells with the immune system. This knowledge is continuously translated into novel treatment concepts and targets, which are dynamically reshaping the therapeutic landscape of these tumors. In this review, we provide a concise overview of the etiology and molecular pathology of the six most common cancers of the digestive system, including esophageal, gastric, biliary tract, pancreatic, hepatocellular, and colorectal cancers. We comprehensively describe the current stage-dependent pharmacological management of these malignancies, including chemo-, targeted, and immunotherapy. For each cancer entity, we provide an overview of recent therapeutic advancements and research progress. Finally, we describe how novel insights into tumor heterogeneity and immune evasion deepen our understanding of therapy resistance and provide an outlook on innovative therapeutic strategies that will shape the future management of digestive cancers, including CAR-T cell therapy, novel antibody-drug conjugates and targeted therapies.
Collapse
Affiliation(s)
- Tianzuo Zhan
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johannes Betge
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Schulte
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Dreikhausen
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Hirth
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Moying Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philip Weidner
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Antonia Leipertz
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Teufel
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany.
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
134
|
Luo YG, Wu M, Chen HG. Retrospective analysis of pathological types and imaging features in pancreatic cancer: A comprehensive study. World J Gastrointest Oncol 2025; 17:99153. [PMID: 39817138 PMCID: PMC11664627 DOI: 10.4251/wjgo.v17.i1.99153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Pancreatic cancer remains one of the most lethal malignancies worldwide, with a poor prognosis often attributed to late diagnosis. Understanding the correlation between pathological type and imaging features is crucial for early detection and appropriate treatment planning. AIM To retrospectively analyze the relationship between different pathological types of pancreatic cancer and their corresponding imaging features. METHODS We retrospectively analyzed the data of 500 patients diagnosed with pancreatic cancer between January 2010 and December 2020 at our institution. Pathological types were determined by histopathological examination of the surgical specimens or biopsy samples. The imaging features were assessed using computed tomography, magnetic resonance imaging, and endoscopic ultrasound. Statistical analyses were performed to identify significant associations between pathological types and specific imaging characteristics. RESULTS There were 320 (64%) cases of pancreatic ductal adenocarcinoma, 75 (15%) of intraductal papillary mucinous neoplasms, 50 (10%) of neuroendocrine tumors, and 55 (11%) of other rare types. Distinct imaging features were identified in each pathological type. Pancreatic ductal adenocarcinoma typically presents as a hypodense mass with poorly defined borders on computed tomography, whereas intraductal papillary mucinous neoplasms present as characteristic cystic lesions with mural nodules. Neuroendocrine tumors often appear as hypervascular lesions in contrast-enhanced imaging. Statistical analysis revealed significant correlations between specific imaging features and pathological types (P < 0.001). CONCLUSION This study demonstrated a strong association between the pathological types of pancreatic cancer and imaging features. These findings can enhance the accuracy of noninvasive diagnosis and guide personalized treatment approaches.
Collapse
Affiliation(s)
- Yang-Gang Luo
- Pathology Department, Xuanhan County People’s Hospital, Dazhou 636150, Sichuan Province, China
| | - Mei Wu
- Pathology Department, Xuanhan County People’s Hospital, Dazhou 636150, Sichuan Province, China
| | - Hong-Guang Chen
- Pathology Department, Xuanhan County People’s Hospital, Dazhou 636150, Sichuan Province, China
| |
Collapse
|
135
|
Wu Q, Xiao Q, Tang X, Li L, Song D, Zhou Y, Li B, Ren G, Luo F. DAMPs prognostic signature predicts tumor immunotherapy, and identifies immunosuppressive mechanism of pannexin 1 channels in pancreatic ductal adenocarcinoma. Front Immunol 2025; 15:1516457. [PMID: 39882247 PMCID: PMC11775746 DOI: 10.3389/fimmu.2024.1516457] [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: 10/24/2024] [Accepted: 12/13/2024] [Indexed: 01/31/2025] Open
Abstract
Background Damage-associated molecular patterns (DAMPs) induced by immunogenic cell death (ICD) may be useful for the immunotherapy to patients undergoing pancreatic ductal adenocarcinoma (PDAC). The aim of this study is to predict the prognosis and immunotherapy responsiveness of PDAC patients using DAMPs-related genes. Methods K-means analysis was used to identify the DAMPs-related subtypes of 175 PDAC cases. The significance of gene mutation and immune status in different subtypes was detected. LASSO regression was used to construct a DAMPs-related prognostic signature to predict the immunotherapy responsiveness of PDAC. Subsequently, in vivo and in vitro experiments and Bulk-RNA seq were used to verify the effect of hub gene pannexin 1 (PANX1) on PDAC. Results Two subtypes were clustered based on the expression levels of DAMPs genes from 175 PDAC patients. Besides, the prognosis and immune landscape in up-regulated DAMPs expression subtypes was poor. In addition, we constructed a DAMPs-related prognostic signature that correlated with immune cell infiltration and predicted immunotherapy or chemotherapy responsiveness of patients with PDAC. Mechanically, through Bulk-RNA sequencing and experiments, we found that PANX1 promoted tumor progression and immune regulation via the ATP release to active NOD1/NFκB signaling pathway in PDAC. Conclusion Our in silico analyses established a classification system based on ICD-related DAMPs genes in PDAC, and constructed a DAMPs-related prognostic model to predict the efficacy of immunotherapy. This study will provide a new perspective for targeting the DAMPs-related molecule PANX1 in the treatment of PDAC.
Collapse
Affiliation(s)
- Qianxue Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Xiao
- Department of Breast and Thyroid Surgery, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Tang
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Chongqing Liang Jiang New Area, Chongqing, China
| | - Liuying Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Daqiang Song
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Benhua Li
- Department of Clinical Laboratory, The Second People’ s Hospital of Liangshan yi Autonomous Prefecture, Xichang, China
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Luo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
136
|
Ding M, Zong Q, Zhang D, Ullah I, Zhang X, Liang W, Li X, Bulatov E, Yuan Y. Self-Adaptive Nanocarriers Overcome Multiple Physiological Barriers to Boosting Chemotherapy of Orthotopic Pancreatic Cancer. ACS NANO 2025; 19:662-679. [PMID: 39731749 DOI: 10.1021/acsnano.4c11514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2024]
Abstract
Chemotherapy is the primary treatment option for pancreatic cancer, although nanocarrier-based drug delivery systems often struggle with multiple physiological barriers, limiting their therapeutic efficacy. Here, we developed a pH/reactive oxygen species (ROS) dual-sensitive self-adaptive nanocarrier (DATCPT) encapsulating camptothecin (CPT), an analog of the pancreatic chemotherapeutic drug irinotecan (CPT-11), to enhance chemotherapy outcomes in orthotopic pancreatic cancer by addressing multiple physiological barriers. The nanocarrier features a peripherally positively charged arginine (Arg) residue on DATCPT and is masked with an acid-labile 2,3-dimethylmaleic anhydride (DA) to improve circulation time. In the acidic tumor microenvironment (TME), DA dissociates, exposing arginine to facilitate nanocarrier binding and internalization of DATCPT. Subsequently, peroxynitrite (ONOO-) is generated by a cascade reaction between exposed Arg and ROS, which effectively activates matrix metalloproteinases (MMPs) to degrade the dense extracellular matrix (ECM) and enhance the deep accumulation and penetration of DATCPT. Meanwhile, ONOO- inhibits tumor metastasis by influencing mitochondrial function, preventing adenosine triphosphate (ATP) production, and inhibiting ATP-dependent tumor-derived microvesicles (TMVs). This study presents a promising strategy to develop efficient nanocarriers to address multiple physiological barriers in antipancreatic cancer therapy.
Collapse
Affiliation(s)
- Mengchao Ding
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
| | - Qingyu Zong
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Dan Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P. R. China
| | - Ihsan Ullah
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P. R. China
| | - Xingzu Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P. R. China
| | - Wenhua Liang
- Department of Radiology, First Affiliated Hospital of the Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, P. R. China
| | - Xinchun Li
- Department of Radiology, First Affiliated Hospital of the Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, P. R. China
| | - Emil Bulatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Youyong Yuan
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| |
Collapse
|
137
|
Yao H, Ren Y, Wu F, Cao L, Liu J, Yan M, Li X. The Discovery of a Novel AXL/Triple Angiokinase Inhibitor Based on 6-Chloro-Substituted Indolinone and Side Chain Methyl Substitution Inhibiting Pancreatic Cancer Growth and Metastasis. J Med Chem 2025; 68:465-490. [PMID: 39711508 DOI: 10.1021/acs.jmedchem.4c02130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
In this study, we discovered and identified a novel AXL/triple angiokinase inhibitor 11b by rational structural modification based on the structure of triple angiokinase inhibitor Nintedanib. We found that 11b potently inhibited AXL expression with the IC50 value of 3.75 nM and possessed similar inhibitory activity on KDR as Nintedanib. In the assay of antiproliferative activity on NIH/3T3, HUVEC, Bxpc-3, and MDA-MB-231, 11b showed better inhibitory ability than Nintedanib. In pancreatic cancer xenograft mouse models from Bxpc-3 cells, even when the dosage was halved, 11b exhibited better or comparable effects to Nintedanib (tumor growth inhibition (TGI) based on tumor volume change during the trial or tumor weight). Notably, we also found that 11b prohibited Bxpc-3 resulted lung metastasis by inhibiting its epithelial-mesenchymal transition (EMT) process. Another mechanism assay also proved that 11b inhibited the function of blood vessels and fibroblasts, promoted apoptosis of cancer and fibroblast cells, and exhibited low toxicity and good metabolic stability.
Collapse
Affiliation(s)
- Han Yao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuanyuan Ren
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Feng Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Longcai Cao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
| | - Jiadai Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Ming Yan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xingshu Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
138
|
Wang X, Yu P, Jia W, Wan B, Ling Z, Tang Y. Integrating traditional machine learning with qPCR validation to identify solid drug targets in pancreatic cancer: a 5-gene signature study. Front Pharmacol 2025; 15:1539120. [PMID: 39850570 PMCID: PMC11754184 DOI: 10.3389/fphar.2024.1539120] [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: 12/03/2024] [Accepted: 12/20/2024] [Indexed: 01/25/2025] Open
Abstract
Background Pancreatic cancer remains one of the deadliest malignancies, largely due to its late diagnosis and lack of effective therapeutic targets. Materials and methods Using traditional machine learning methods, including random-effects meta-analysis and forward-search optimization, we developed a robust signature validated across 14 publicly available datasets, achieving a summary AUC of 0.99 in training datasets and 0.89 in external validation datasets. To further validate its clinical relevance, we analyzed 55 peripheral blood samples from pancreatic cancer patients and healthy controls using qPCR. Results This study identifies and validates a novel five-gene transcriptomic signature (LAMC2, TSPAN1, MYO1E, MYOF, and SULF1) as both diagnostic biomarkers and potential drug targets for pancreatic cancer. The differential expression of these genes was confirmed, demonstrating their utility in distinguishing cancer from normal conditions with an AUC of 0.83. These findings establish the five-gene signature as a promising tool for both early, non-invasive diagnostics and the identification of actionable drug targets. Conclusion A five-gene signature is established robustly and has utility in diagnostics and therapeutic targeting. These findings lay a foundation for developing diagnostic tests and targeted therapies, potentially offering a pathway toward improved outcomes in pancreatic cancer management.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Pengcheng Yu
- Department of General Surgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Wei Jia
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bingbing Wan
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhougui Ling
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Yangyang Tang
- Department of General Surgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| |
Collapse
|
139
|
Aseafan M, Alfakeeh AH, Tashkandi E, Mahrous M, Alghamdi M, Alshamsan B, Al-Hajeili M, Bakhsh S, Alshammari K, Almugbel FA, Alfagih AH, Allehebi A, Montaser M, Elsafty MH, Elnaghi KAE, Issa I, Bakshi E, AlSubaie S, AlMutairi B, Mokhtar H, Aboelatta M, Bukhari N, Alzahrani AM, Elhassan T, Alqahtani A, Bazarbashi S. Real-world clinical outcome of unresectable locally advanced & de-novo metastatic pancreatic ductal adenocarcinoma: a multicentre retrospective study. BMC Cancer 2025; 25:7. [PMID: 39754118 PMCID: PMC11697791 DOI: 10.1186/s12885-024-13386-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with limited treatment options yielding poor outcomes. This study aimed to evaluate the real-world clinical characteristics, treatment patterns, and outcomes of patients with locally advanced unresectable and de-novo metastatic PDAC in Saudi Arabia, providing regional data to compare with international benchmarks. METHODS This is a retrospective, multicentre study involving 350 patients diagnosed with unresectable locally advanced or de-novo metastatic PDAC between January 2015 and November 2023. Data were collected from 10 oncology centers across Saudi Arabia. RESULTS The median age at diagnosis was 60 years, with 63% of patients presenting with multiple metastatic sites, primarily in the liver (66.3%). FOLFIRINOX was the most common first-line treatment (55.1%), followed by gemcitabine plus nab-paclitaxel (15.1%). The median PFS for first-line treatment was 5.3 months, with FOLFIRINOX achieving the longest PFS (6.5 months). The median OS was 10.34 months for the entire cohort, with better survival outcomes observed in patients receiving FOLFIRINOX (12.3 months). Independent prognostic factors for PFS and OS included performance status, first-line regimen, and neutrophil-lymphocyte ratio (NLR). Among patients tested, 7.1% had deficient mismatch repair (d-MMR), and 5.8% harbored BRCA mutations. CONCLUSIONS This real-world study confirms that clinical outcomes for locally advanced unresectable and metastatic PDAC in Saudi Arabia are consistent with international data, with FOLFIRINOX showing superior outcomes over gemcitabine-based regimens. However, both treatments reflect the persistent poor prognosis of PDAC, underscoring the need for novel therapeutic strategies. Further research is warranted to optimize treatment selection and improve survival outcomes in this population.
Collapse
Affiliation(s)
- Mohamed Aseafan
- Section of Medical Oncology, Department of Internal Medicine, Security Forces Hospital, Riyadh, Saudi Arabia.
| | - Ali H Alfakeeh
- Comprehensive Cancer Center, Medical Oncology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Emad Tashkandi
- College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mervat Mahrous
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
- Collage of Medicine, Minia University, Minia, Egypt
| | - Mohammed Alghamdi
- Oncology Center, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Bader Alshamsan
- Department of Medicine, College of Medicine, Qassim University, Buraydah, Saudi Arabia
- Prince Faisal Cancer Center, King Fahad Specialist Hospital, Qassim Health Clusster, Buraydah, Saudi Arabia
| | - Marwan Al-Hajeili
- Department of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Safwan Bakhsh
- Department of Medical Oncology, King Faisal Specialist Hospital and Research Center-Jeddah, Jeddah, Saudi Arabia
| | - Kanan Alshammari
- Department of Medical Oncology, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Fahad A Almugbel
- Department of Medical Oncology, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Abdulhameed H Alfagih
- Comprehensive Cancer Center, Medical Oncology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ahmed Allehebi
- Department of Medical Oncology, King Faisal Specialist Hospital and Research Center-Jeddah, Jeddah, Saudi Arabia
| | - Mohamed Montaser
- Section of Medical Oncology, Department of Internal Medicine, Security Forces Hospital, Riyadh, Saudi Arabia
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | - Ibrahim Issa
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Eesa Bakshi
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Sadeem AlSubaie
- Pathology and Laboratory Medicine, Security Forces Hospital, Riyadh, Saudi Arabia
| | - Bandar AlMutairi
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Hoda Mokhtar
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Mohamed Aboelatta
- Prince Faisal Cancer Center, King Fahad Specialist Hospital, Qassim Health Clusster, Buraydah, Saudi Arabia
| | - Nedal Bukhari
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Ali M Alzahrani
- Comprehensive Cancer Center, Medical Oncology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Tusneem Elhassan
- Research Unit, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ali Alqahtani
- Department of Medical Oncology, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Shouki Bazarbashi
- Department of Medical Oncology, Cancer Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
140
|
Chung TT, Piao Z, Lee SJ. Identification of ferroptosis-related signature predicting prognosis and therapeutic responses in pancreatic cancer. Sci Rep 2025; 15:75. [PMID: 39748113 PMCID: PMC11695983 DOI: 10.1038/s41598-024-84607-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025] Open
Abstract
Ferroptosis plays a role in tumorigenesis by affecting lipid peroxidation and metabolic pathways; however, its prognostic or therapeutic relevance in pancreatic adenocarcinoma (PAAD) remains poorly understood. In this study, we developed a prognostic ferroptosis-related gene (FRG)-based risk model using cohorts of The Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC), proposing plausible therapeutics. Differentially expressed FRGs between tumors from TCGA-PAAD and normal pancreatic tissues from Genotype-Tissue Expression were analyzed to construct a prognostic risk model using univariate and multivariate Cox regression and LASSO analyses. A model incorporating AURKA, CAV1, and PML gene expression effectively distinguished survival differences between high- and low-risk groups among TCGA-PAAD patients, with validation in two ICGC cohorts. The high-risk group was enriched in gene sets involving mTOR, MAPK, and E2F signaling. The immune and stromal cells infiltration score did not differ between the groups. Analysis of PRISM datasets using our risk model to classify pancreatic cell lines suggested the dasatinib's efficacy in the high-risk group, which was experimentally confirmed in four cell lines with a high- or low-risk signature. In conclusion, this study proposed a robust FRG-based prognostic model that may help stratify PAAD patients with poor prognoses and select potential therapeutic avenues.
Collapse
Affiliation(s)
- Ting Ting Chung
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Zanyue Piao
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Seung Jin Lee
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
141
|
Peng J, Sun J, Yu Y, Yuan Q, Zhang Y. Integrative multi-omics analysis reveals the role of toll-like receptor signaling in pancreatic cancer. Sci Rep 2025; 15:52. [PMID: 39747201 PMCID: PMC11696379 DOI: 10.1038/s41598-024-84062-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
As one of the most destructive and invasive cancers, pancreatic cancer exhibits complex tumor heterogeneity, which has been a major challenge for clinicians in terms of patient treatment and prognosis. The toll-like receptor (TLR) pathway is closely related to the immune microenvironment within various cancer tissues. To explore the development pattern of pancreatic cancer and find an ideal biomarker, our research has explored the mechanism of the TLR pathway in pancreatic cancer. We collected single-cell expression data from 57,024 cells and transcriptomic data from 945 pancreatic cancer patients, and conducted a series of analyses at both the single-cell and transcriptomic levels. By calculating the TLR pathway score, we clustered pancreatic cancer patients and conducted a series of analyses including metabolic pathways, immune microenvironment, drug sensitivity and so on. In the process of building prognostic models, we screened 33 core genes related to the prognosis of pancreatic cancer, and combined a series of machine learning algorithms to build the prognosis model of pancreatic cancer. We used single cell sequencing to clarify the complex intrinsic relationship between TLR pathway and pancreatic cancer. The strongest TLR signals were observed in macrophages and endothelial cells. With the occurrence of pancreatic cancer, the TLR signal of various cell types gradually increased, but with the increase of the malignant degree of ductal epithelial cells, the TLR signal gradually weakened. Cluster analysis showed that patients with the most active TLR pathway had severe dysregulation of immune microenvironment and the worst prognosis. Finally, we combined a series of machine learning algorithms to build a pancreatic cancer prognosis model that includes four genes (NT5E, TGFBI, ANLN, and FAM83A). The model showed strong performance in predicting the survival state of pancreatic cancer samples. We explored the important role of TLR pathway in pancreatic cancer and established and validated a new prognosis model for pancreatic cancer based on TLR-related genes.
Collapse
Affiliation(s)
- Jie Peng
- Ningde Clinical Medical College of Fujian Medical University, Fujian, China
- Ningde Municipal Hospital of Ningde Normal University, Fujian, China
| | - Jiaao Sun
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Youfeng Yu
- Ningde Clinical Medical College of Fujian Medical University, Fujian, China
- Ningde Municipal Hospital of Ningde Normal University, Fujian, China
| | - Qihang Yuan
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Yong Zhang
- Ningde Clinical Medical College of Fujian Medical University, Fujian, China.
- Ningde Municipal Hospital of Ningde Normal University, Fujian, China.
| |
Collapse
|
142
|
Zhang J, Wang J, Li Y, Zheng Y, Hai P, Zhang J. Highly specific GSH-triggered bifunctional molecules to enable precise imaging and targeted therapy of cancer. Talanta 2025; 281:126862. [PMID: 39265421 DOI: 10.1016/j.talanta.2024.126862] [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: 07/21/2024] [Revised: 09/01/2024] [Accepted: 09/07/2024] [Indexed: 09/14/2024]
Abstract
The utilization of diagnostic-integrated molecules can enable targeted delivery and controlled release to significantly enhance therapeutic effectiveness and minimize toxic effects. Herein, we developed a novel class of glutathione (GSH)-activated bifunctional molecules that respond to elevated levels of GSH in tumor microenvironment. These bifunctional molecules retained the pharmacodynamic effects of parent molecules and mitigated cytotoxicity. Meanwhile, controlled release was monitored using fluorescent signals, enabling detection of drug distribution and accumulation in situ and in real time. Moreover, the correlation between GSH levels and fluorescence intensity offers the possibility of monitoring the effectiveness of responsive drugs. In conclusion, bifunctional molecules, as novel diagnostic-integrated molecules with both fluorescence imaging and therapeutic effects, exhibited potential applications in cancer therapy and imaging.
Collapse
Affiliation(s)
- Junyu Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jin Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yanchen Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yongbiao Zheng
- NMPA Key Laboratory for Quality Control of Traditional Chinese and Tibetan Medicine, Qinghai Provincial Drug Inspection and Testing Institute, Xining, 810016, China
| | - Ping Hai
- NMPA Key Laboratory for Quality Control of Traditional Chinese and Tibetan Medicine, Qinghai Provincial Drug Inspection and Testing Institute, Xining, 810016, China
| | - Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
143
|
Adachi T, Nakamura M, Iwai T, Yoshimura M, Mizowaki T. Delta-Radiomics Approach Using Contrast-Enhanced and Noncontrast-Enhanced Computed Tomography Images for Predicting Distant Metastasis in Patients With Borderline Resectable Pancreatic Carcinoma. Adv Radiat Oncol 2025; 10:101669. [PMID: 39687476 PMCID: PMC11647472 DOI: 10.1016/j.adro.2024.101669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 10/21/2024] [Indexed: 12/18/2024] Open
Abstract
Purpose To predict distant metastasis (DM) in patients with borderline resectable pancreatic carcinoma using delta-radiomics features calculated from contrast-enhanced computed tomography (CECT) and non-CECT images. Methods and Materials Among 250 patients who underwent radiation therapy at our institution between February 2013 and December 2021, 67 patients were deemed eligible. A total of 11 clinical features and 3906 radiomics features were incorporated. Radiomics features were extracted from CECT and non-CECT images, and the differences between these features were calculated, resulting in delta-radiomics features. The patients were randomly divided into the training (70%) and test (30%) data sets for model development and validation. Predictive models were developed with clinical features (clinical model), radiomics features (radiomics model), and a combination of the abovementioned features (hybrid model) using Fine-Gray regression (FG) and random survival forest (RSF). Optimal hyperparameters were determined using stratified 5-fold cross-validation. Subsequently, the developed models were applied to the remaining test data sets, and the patients were divided into high- or low-risk groups based on their risk scores. Prognostic power was assessed using the concordance index, with 95% CIs obtained through 2000 bootstrapping iterations. Statistical significance between the above groups was assessed using Gray's test. Results At a median follow-up period of 23.8 months, 47 (70.1%) patients developed DM. The concordance indices of the FG-based clinical, radiomics, and hybrid models were 0.548, 0.603, and 0.623, respectively, in the test data set, whereas those of the RSF-based models were 0.598, 0.680, and 0.727, respectively. The RSF-based model, including delta-radiomics features, significantly divided the cumulative incidence curves into two risk groups (P < .05). The feature map of the gray-level size-zone matrix showed that the difference in feature values between CECT and non-CECT images correlated with the incidence of DM. Conclusions Delta-radiomics features obtained from CECT and non-CECT images using RSF successfully predict the incidence of DM in patients with borderline resectable pancreatic carcinoma.
Collapse
Affiliation(s)
- Takanori Adachi
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Mitsuhiro Nakamura
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
- Department of Advanced Medical Physics, Graduate School of Medicine, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Takahiro Iwai
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Michio Yoshimura
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Takashi Mizowaki
- Department of Radiation Oncology and Image-Applied Therapy, Kyoto University, Shogoin, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
144
|
Qian X, Liu Y, Chen W, Zheng S, Lu Y, Qiu P, Ke X, Tang H, Zhang X. Paris saponin VII induces Caspase-3/GSDME-dependent pyroptosis in pancreatic ductal adenocarcinoma cells by activating ROS/Bax signaling. CHINESE HERBAL MEDICINES 2025; 17:94-107. [PMID: 39949804 PMCID: PMC11814252 DOI: 10.1016/j.chmed.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/14/2024] [Accepted: 04/07/2024] [Indexed: 02/16/2025] Open
Abstract
Objective Paridis Rhizoma (Chonglou in Chinese), a traditional Chinese herbal medicine, has been shown have strong anti-tumor effects. Paris saponin VII (PSVII), an active constituent isolated from Paridis Rhizoma, was demonstrated to significantly suppress the proliferation of BxPC-3 cells in our previous study. Here, we aimed to elucidate the anti-pancreatic ductal adenocarcinoma (PDAC) effect of PSVII and the underlying mechanism. Methods Cell viability was determined by CCK-8, colony formation, and cell migration assays. Cell apoptosis and reactive oxygen species (ROS) production were measured by flow cytometry with annexin V/propidine iodide (Annexin V/PI) and 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA), respectively. Pyroptosis was evaluated by morphological features, Hoechst 33342/PI staining assay, and release of lactate dehydrogenase (LDH). JC-1 fluorescent dye was employed to measure mitochondrial membrane potential. Western blotting and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to determine the levels of proteins or mRNAs. The effect in vivo was assessed by a xenograft tumor model. Results PSVII inhibited the viability of PDAC cells (BxPC-3, PANC-1, and Capan-2 cells) and induced gasdermin E (GSDME) cleavage, as well as the simultaneous cleavage of Caspase-3 and poly (ADP-ribose) polymerase 1 (PARP). Knockdown of GSDME shifted PSVII-induced pyroptosis to apoptosis. Additionally, the effect of PSVII was significantly attenuated by Z-Asp(OMe)-Glu(OMe)-Val-Asp(OMe)-fluoromethylketone (Z-DEVD-FMK), on the induction of GSDME-dependent pyroptosis. PSVII also elevated intracellular ROS accumulation and stimulated Bax and Caspase-3/GSDME to conduct pyroptosis in PDAC cells. The ROS scavenger N-acetyl cysteine (NAC) suppressed the release of LDH and inhibited Caspase-9, Caspase-3, and GSDME cleavage in PDAC cells, ultimately reversing PSVII-induced pyroptosis. Furthermore, in a xenograft tumor model, PSVII markedly suppressed the growth of PDAC tumors and induced pyroptosis. Conclusion These results demonstrated that PSVII exerts therapeutic effects through Caspase-3/GSDME-dependent pyroptosis and may constitute a novel strategy for preventing chemotherapeutic resistance in patients with PDAC in the future.
Collapse
Affiliation(s)
- Xiaoying Qian
- Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an 710032, China
| | - Yang Liu
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an 710032, China
| | - Wenwen Chen
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an 710032, China
| | - Shuxian Zheng
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Yunyang Lu
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an 710032, China
| | - Pengcheng Qiu
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an 710032, China
| | - Xisong Ke
- Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Haifeng Tang
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an 710032, China
| | - Xue Zhang
- Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
145
|
Shahbaz M, Perween A, Momal U, Imran M, Ul Hassan MH, Naeem H, Mujtaba A, Hussain M, Alsagaby SA, Al Abdulmonem W, Abdelgawad MA, El‐Ghorab AH, Selim S, Mostafa EM, Al Jbawi E. Recent Perspectives on Anticancer Potential of Coumarin Against Different Human Malignancies: An Updated Review. Food Sci Nutr 2025; 13:e4696. [PMID: 39803273 PMCID: PMC11717051 DOI: 10.1002/fsn3.4696] [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: 08/21/2024] [Revised: 10/23/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025] Open
Abstract
Coumarins, a group of naturally occurring compounds, have been reported to demonstrate anticancer potential. These substances, distinguished by their combined benzene and α-pyrone rings, have been demonstrated to impact multiple cellular mechanisms essential for the initiation and advancement of cancer. These agents work in different ways that prevent different tumor cells from growing, spreading, and increasing. One of the main anticancer mechanisms of coumarin act is killing cancer cells through apoptosis. This includes changes to pro- and anti-apoptotic proteins like Bcl-2 and Bax, the release of cytochrome c from mitochondria, and the activation of caspases. The tumor suppressor protein p53's expression has been discovered to be upregulated by coumarins such as esculetin and imperatorin, which encourage interrupted cell cycle and death. Additionally, coumarin has anti-angiogenic qualities, which are critical for the development and spread of tumors. It can slow the development of new blood vessels that feed tumors by inhibiting the "vascular endothelial growth factor (VEGF)" route of signaling. Coumarins inhibit the number of signaling pathways that are vital for cell division. For example, they can suppress the "PI3K/mTOR" pathway, which usually impairs the cancer cells and results in decreased cell viability and growth. Finally, coumarins could modulate the response of the immune system to cancerous cells. They have the ability to boost the activity of natural killer cells and cytotoxic T lymphocytes, which aid the immune system in identifying and eliminating cancer cells. Through a variety of mechanisms, such as immune response regulation, angiogenesis reduction, cell growth inhibition, and apoptosis activation, coumarins exhibit their anticancer effects. These molecular pathways demonstrate coumarins' potential as an interesting option for the development of novel anticancer treatments. More studies are needed to completely understand their modes of action and maximize their therapeutic efficacy.
Collapse
Affiliation(s)
- Muhammad Shahbaz
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of Agriculture MultanMultanPakistan
| | - Asfa Perween
- Department of Human Nutrition and DieteticsMuhammad Nawaz Shareef University of Agriculture MultanMultanPakistan
| | - Ushna Momal
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of Agriculture MultanMultanPakistan
| | - Muhammad Imran
- Department of Food Science and TechnologyUniversity of NarrowalNarrowalPakistan
| | - Muhammad Hammad Ul Hassan
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of Agriculture MultanMultanPakistan
| | - Hammad Naeem
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of Agriculture MultanMultanPakistan
- Post Harvest Research CentreAyub Agricultural Research Institute FaisalabadFaisalabadPakistan
| | - Ahmed Mujtaba
- Department of Food Science and Technology, Faculty of Engineering Sciences and TechnologyHamdard University Islamabad CampusIslamabadPakistan
| | - Muzzamal Hussain
- Department of Food SciencesGovernment College University FaisalabadFaisalabadPakistan
| | - Suliman A. Alsagaby
- Department of Medical Laboratory Sciences, College of Applied Medical SciencesMajmaah UniversityAl‐MajmaahSaudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of MedicineQassim UniversityBuraidahSaudi Arabia
| | - Mohamed A. Abdelgawad
- Department of Pharmaceutical Chemistry, College of PharmacyJouf UniversitySakakaAljoufSaudi Arabia
| | - Ahmed H. El‐Ghorab
- Department of Chemistry, College of ScienceJouf UniversitySakakaSaudi Arabia
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesJouf UniversitySakakaSaudi Arabia
| | - Ehab M. Mostafa
- Department of Pharmacognosy, College of PharmacyJouf UniversitySakakaSaudi Arabia
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys)Al‐Azhar UniversityCairoEgypt
| | | |
Collapse
|
146
|
Cheng L, Wei L, Chen Q, Li P, Zhang D. DDR1 in pancreatic adenocarcinoma: unraveling the mechanisms of immune exclusion. Scand J Gastroenterol 2025; 60:81-90. [PMID: 39713856 DOI: 10.1080/00365521.2024.2443505] [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: 11/21/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAAD) is a deadly cancer marked by extensive collagen deposition and limited response to immunotherapy. Discoidin domain receptor1 (DDR1), part of the transmembrane receptor tyrosine kinase family, is linked to inflammation regulation and immune cell infiltration. However, its role in controlling cytokines and chemokines in the microenvironment of PAAD is still unclear. METHODS Initially, RNA sequencing data from TCGA were utilized to investigate the expression of DDR1. Subsequently, the relationship between DDR1 and immune infiltration was examined through Gene Ontology (GO) and Gene Set Enrichment Analysis (GSEA) analysis, in conjunction with ssGSEA Immunoanalysis. Lastly, the effect of DDR1 on the malignant characteristics of PAAD cells was examined through in vitro experimentation, employing various techniques such as the CCK8 assay, colony formation assay and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). RESULTS The research revealed a notable increase in the expression level of DDR1 in PAAD. Subsequent analysis indicated a correlation between the differential expression of DDR1 with chemokines and immune infiltration. Additionally, cellular experiments demonstrated that the downregulation of DDR1 led to enhanced expression of chemokines CCL4, CCL5 and CXCL10. CONCLUSION DDR1 is linked to tumor immune infiltration, and the knockout of DDR1 results in the upregulation of chemokines CCL4, CCL5 and CXCL10 in Pan02 PAAD cells.
Collapse
Affiliation(s)
- Long Cheng
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lina Wei
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Qian Chen
- The Second Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Peirong Li
- Department of Emergency, Lanzhou University Second Hospital, Lanzhou, China
| | - Dekui Zhang
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
147
|
Hu Y, Liao Y, Pan S, Zhou J, Wan C, Huang F, Bai Y, Lin C, Xia Q, Liu Z, Gong J, Nie X, Wang M, Qin R. A Triple-Mismatch Differentiating assay exploiting activation and trans cleavage of CRISPR-Cas12a for mutation detection with ultra specificity and sensitivity. Biosens Bioelectron 2025; 267:116826. [PMID: 39369517 DOI: 10.1016/j.bios.2024.116826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/22/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Liquid biopsy technology is non-invasive and convenient, and is currently an emerging technology for cancer screening. Among them, clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR associated protein 12a (Cas12a) based nucleic acid detection technology has the advantages of high sensitivity, rapidity, and easy operation. However, CRISPR-Cas12a does not discriminate single-base mismatches of targets well enough to meet the needs of clinical detection. Herein, we developed the Triple-Mismatch Differentiating (TMD) assay. This assay amplified the small thermodynamic difference in mismatches at one site at the level of CRISPR-Cas12a activation to a significant thermodynamic difference at three sites at both the level of CRISPR-Cas12a activation and trans-cleavage, which greatly improves the ability of CRISPR-Cas12a to discriminate between base mismatches. Our manipulation greatly improved the specificity of the CRISPR-Cas12a system while maintaining its inherent sensitivity and simplicity, increasing the detection limit to 0.0001%. When testing samples from pancreatic cancer patients, our results were highly consistent with NGS sequencing results. We believe that the TMD assay will provide a new technology for early cancer detection and will be widely used in the clinical practice.
Collapse
Affiliation(s)
- Yibo Hu
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yangwei Liao
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Shutao Pan
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jingcong Zhou
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Changqing Wan
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Feiyang Huang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yu Bai
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Chen Lin
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Qilong Xia
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zixi Liu
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jun Gong
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Xiaoqi Nie
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
148
|
Duran T, Balikci I, Buyukkosucu B, Gunes IF, Pekgonul HK, Vardar N, Yilmaz MD, Ak G, Zengin G. Biological Characterization of One Oxadiazole Derivative (5(4-Hydroxyphenyl)-2-(N-Phenyl Amino)-1,3,4-Oxadiazole): In Vitro, In Silico, and Network Pharmacological Approaches. Chem Biol Drug Des 2025; 105:e70038. [PMID: 39757393 DOI: 10.1111/cbdd.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/28/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
Oxadiazole compounds are of great interest because they have a range of biological activities ranging from antioxidants to anticancer agents. Against this background, we wanted to demonstrate the antioxidant, enzyme inhibitory, and anticancer effects of 5(4-hydroxyphenyl)-2-(N-phenylamino)-1,3,4-oxadiazole (Hppo). Antioxidant abilities were measured through free radical scavenging and reducing power tests. Enzyme inhibitory effects were studied by cholinesterases, tyrosinase, amylase, and glucosidase. The anticancer effect was tested on pancreatic cancer cell lines (PANC-1, CRL-169) and on HEK293 cell lines. The compound showed significant antioxidant activity (particularly in the CUPRAC (cupric acid-reducing antioxidant capacity) assay) and enzyme inhibitory properties (particularly glucosidase inhibition). In the anticancer test, the compound showed strong anticancer activity in pancreatic cancer with apoptotic signaling pathways. These results were confirmed by molecular modeling and bioinformatics tools. Thus, our findings can provide novel and versatile compounds for the development of multidirectional drugs in the pharmaceutical industry.
Collapse
Affiliation(s)
- Tugce Duran
- Department of Medical Genetics, Faculty of Medicine, KTO Karatay University, Konya, Turkey
- Department of Pediatric Allergy and Immunology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Irem Balikci
- Department of Materials Science and Nanotechnology Engineering, Faculty of Engineering and Natural Sciences, KTO Karatay University, Konya, Turkey
| | - Busra Buyukkosucu
- Department of Materials Science and Nanotechnology Engineering, Faculty of Engineering and Natural Sciences, KTO Karatay University, Konya, Turkey
| | - Ibrahim Furkan Gunes
- Department of Materials Science and Nanotechnology Engineering, Faculty of Engineering and Natural Sciences, KTO Karatay University, Konya, Turkey
| | - Hatice Kubra Pekgonul
- Department of Materials Science and Nanotechnology Engineering, Faculty of Engineering and Natural Sciences, KTO Karatay University, Konya, Turkey
| | - Necati Vardar
- Department of Materials Science and Nanotechnology Engineering, Faculty of Engineering and Natural Sciences, KTO Karatay University, Konya, Turkey
- Department of Metallurgy and Materials Engineering, Faculty of Engineering and Natural Sciences, KTO Karatay University, Konya, Turkey
| | - Mahmut Deniz Yilmaz
- Department of Basic Sciences, Faculty of Engineering, Necmettin Erbakan University, Konya, Turkey
- BITAM-Science and Technology Research and Application Center, Necmettin Erbakan University, Konya, Turkey
| | - Gunes Ak
- Department of Biology, Faculty of Science, Selcuk University, Konya, Turkey
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University, Konya, Turkey
| |
Collapse
|
149
|
Malhotra K, Malik A, Almalki WH, Sahebkar A, Kesharwani P. Reactive Oxygen Species and its Manipulation Strategies in Cancer Treatment. Curr Med Chem 2025; 32:55-73. [PMID: 37303173 DOI: 10.2174/0929867330666230609110455] [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: 04/01/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023]
Abstract
Cancer is one of the serious diseases of modern times, occurring in all parts of the world and shows a wide range of effects on the human body. Reactive Oxygen Species (ROS) such as oxide and superoxide ions have both advantages and disadvantages during the progression of cancer, dependent on their concentration. It is a necessary part of the normal cellular mechanisms. Changes in its normal level can cause oncogenesis and other relatable problems. Metastasis can also be controlled by ROS levels in the tumor cells, which can be prevented by the use of antioxidants. However, ROS is also used for the initiation of apoptosis in cells by different mediators. There exists a cycle between the production of oxygen reactive species, their effect on the genes, role of mitochondria and the progression of tumors. ROS levels cause DNA damage by the oxidation process, gene damage, altered expression of the genes and signalling mechanisms. They finally lead to mitochondrial disability and mutations, resulting in cancer. This review summarizes the important role and activity of ROS in developing different types of cancers like cervical, gastric, bladder, liver, colorectal and ovarian cancers.
Collapse
Affiliation(s)
- Kabil Malhotra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Arzoo Malik
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| |
Collapse
|
150
|
Zhu Q, Chen Z, Tian M, Yan X, Gongye X, Liu Z, Zhao A, Yang Z, Yuan Y. Improved Predictability of Diagnosis and Prognosis Using Serum- and Tissue-Derived Extracellular Vesicles From Bulk mRNA Sequencing in Pancreatic Ductal Adenocarcinoma. Cancer Med 2025; 14:e70538. [PMID: 39812156 PMCID: PMC11733676 DOI: 10.1002/cam4.70538] [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: 11/09/2023] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Early-stage pancreatic ductal adenocarcinoma (PDAC) is frequently misdiagnosed, contributing to its high mortality rate. Exosomal microRNAs (miRNAs) have emerged as potential biomarkers for the early detection of PDAC. AIMS This study aimed to evaluate the feasibility of using exosomal miRNAs from PDAC tissues and serum as biomarkers for early detection and prognosis. MATERIALS & METHODS Exosomes were isolated from healthy individuals and PDAC patients via tissue and serum samples, then identified by analyzing their particle size and protein content. PDAC-specific exosomal miRNAs were identified using a microRNA array. A large cohort was subsequently recruited to validate these findings. The diagnostic capacity of the identified miRNAs was assessed using the Brier score and area under the curve (AUC). Verified miRNAs were also used to confirm intracellular mRNA change patterns. RESULTS The combination of miR142-3p, miR148a-3p, and CA199 showed a higher AUC (0.747) compared to CA199 alone (0.716) in ROC curve analysis. Gene Ontology (GO) annotations revealed that the two-miRNA panel was associated with multiple oncogenic pathways. DISCUSSION 142-3p and miR148a-3p were identified as specific to PDAC and, when combined with CA199, improved diagnostic accuracy. Their involvement in oncogenic pathways underscores their relevance as diagnostic and prognostic biomarkers. CONCLUSION MiR142-3p and miR148a-3p, alongside CA199, show promise as non-invasive biomarkers for early detection and prognosis of PDAC, improving diagnostic accuracy.
Collapse
Affiliation(s)
- Qian Zhu
- Department of Hepatobiliary and Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei ProvinceWuhanChina
| | - Zhang Chen
- Department of Hepatobiliary and Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Ming Tian
- Department of Hepatobiliary and Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei ProvinceWuhanChina
| | - Xin Yan
- Department of Hepatobiliary and Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xiangdong Gongye
- Department of Hepatobiliary and Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Zhicheng Liu
- Department of Hepatobiliary and Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Anbang Zhao
- Department of Hepatobiliary and Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Zhiyong Yang
- Department of Hepatobiliary and Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei ProvinceWuhanChina
| | - Yufeng Yuan
- Department of Hepatobiliary and Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei ProvinceWuhanChina
- TaiKang Center for Life and Medical SciencesWuhan UniversityWuhanChina
| |
Collapse
|