1
|
Huang X, Wen Z, Cai H, Yu D. The role of quercetin in modulating lipid metabolism and enhancing chemotherapy via the STAT3-CPT1B pathway in pancreatic cancer. Biochem Biophys Res Commun 2025; 772:152033. [PMID: 40412371 DOI: 10.1016/j.bbrc.2025.152033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Pancreatic cancer is a highly aggressive gastrointestinal tumor with limited treatment options, such as surgery and chemotherapy. Thus, further research into its pathogenesis and new treatments is necessary. METHODS Fluorescence-activated cell sorting was employed to sort pancreatic cancer stem cells (PCSCs). Sphere formation assays and Cell Counting Kit-8 (CCK-8) assays were conducted to assess stemness and proliferation capacity. Quantitative real-time PCR and Western blot analysis were employed to assess gene expression levels. Furthermore, immunofluorescence microscopy and chromatin immunoprecipitation assays were conducted to examine alterations in signaling pathways and gene expression. RESULTS Quercetin and gemcitabine may inhibit PANC-1 cells and PCSCs by affecting energy metabolism. Chromatin immunoprecipitation assays revealed an interaction between STAT3 and CPT1B in PCSCs. Quercetin and gemcitabine might affect energy metabolism by inhibiting STAT3 and CPT1B. Manipulating STAT3 expression (overexpression plasmids and siRNA knockdown) altered CPT1B mRNA and protein expression. Although acetyl-CoA reversed the quercetin- and gemcitabine-induced expression of N-cadherin, DECR1, and ALDH, it had minimal influence on CPT1B and STAT3 levels. CONCLUSION Quercetin inhibits the expression of CPT1B via the STAT3 signaling pathway, affecting lipid metabolism and exerting antitumor effects. Furthermore, the combined administration of quercetin and gemcitabine exhibits enhanced therapeutic efficacy.
Collapse
Affiliation(s)
- Xinshi Huang
- Department of Ultrasound, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325003, PR China
| | - Zhengde Wen
- Departments of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325003, PR China
| | - Huajie Cai
- Departments of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325003, PR China
| | - Dinglai Yu
- Departments of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325003, PR China.
| |
Collapse
|
2
|
Li Y, Zhang J, Zheng J, Liu Y, Zhao L, Ma Z. Penpulimab in an adolescent patient with pancreatic neuroendocrine carcinoma with liver metastasis: A case report and literature review. Medicine (Baltimore) 2025; 104:e42730. [PMID: 40489810 DOI: 10.1097/md.0000000000042730] [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] [Indexed: 06/11/2025] Open
Abstract
RATIONALE Pancreatic neuroendocrine carcinoma (PNEC) is a rare, aggressive malignancy with limited therapeutic options and a dismal prognosis, particularly in young patients. Approximately 85% of PNEC cases eventually progress to metastatic cancer. Despite advances in oncology, optimal management strategies for metastatic PNEC remain undefined, especially in chemotherapy-ineligible cases. PATIENT CONCERNS An 18-year-old male patient, reporting anorexia with associated weight loss (15 kg weight loss over 3 months) and significant abdominal distension. DIAGNOSES The results of whole abdominal computed tomography, histopathology, immunohistochemistry, and laboratory examination were consistent with pancreatic neuroendocrine carcinoma with liver metastasis. INTERVENTIONS Due to the family refusal of chemotherapy, and immunohistochemistry revealed programmed death-1 ligand positivity. The patient received penpulimab (anti-programmed death-1) combined with anlotinib capsules and then changed to penpulimab combined with sorafenib capsules after progression. OUTCOMES Initial therapy achieved 13 months of progression-free survival, demonstrating durable disease control. Subsequent progression highlighted challenges of acquired resistance, with no severe treatment-related toxicity. The patient was still alive at the time of follow-up in July 2024. LESSONS Pancreatic neuroendocrine cancer is rare. There are various treatment options available. However, the best treatment plan still needs further exploration. This case underscores that programmed death-1 ligand + PNEC may respond to immunotherapy/antiangiogenic combinations, offering alternatives for chemotherapy-ineligible patients. In addition, young patients with aggressive PNEC represent an understudied population, necessitating tailored strategies.
Collapse
Affiliation(s)
- Yafen Li
- Department of Clinical Pharmacy, Wanbei Coal and Electricity Group General Hospital, Suzhou, Anhui, China
- General Clinical Research Center, Wanbei Coal and Electricity Group General Hospital, Suzhou, Anhui, China
| | - Jing Zhang
- Department of Radiotherapy, Wanbei Coal and Electricity Group General Hospital, Suzhou, Anhui, China
| | - Jie Zheng
- Department of Radiotherapy, Wanbei Coal and Electricity Group General Hospital, Suzhou, Anhui, China
| | - Yujie Liu
- Department of Clinical Pharmacy, Wanbei Coal and Electricity Group General Hospital, Suzhou, Anhui, China
| | - Lu Zhao
- Department of Oncology, Wanbei Coal and Electricity Group General Hospital, Suzhou, Anhui, China
| | - Zhiyu Ma
- Department of Radiotherapy, Wanbei Coal and Electricity Group General Hospital, Suzhou, Anhui, China
| |
Collapse
|
3
|
Ai B, Liang Y, Yan T, Lei Y. Exploration of immune cell heterogeneity by single-cell RNA sequencing and identification of secretory leukocyte protease inhibitor as an oncogene in pancreatic cancer. ENVIRONMENTAL TOXICOLOGY 2025; 40:879-890. [PMID: 38476085 DOI: 10.1002/tox.24200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/08/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024]
Abstract
Clinical outcomes remain unsatisfactory in patients with pancreatic cancer (PAC). In this study, through single-cell sequencing, we identified eight cell subpopulations in the tumor microenvironment (TME). Redimensional clustering of epithelial cells, myeloid cells, and cancer-associated fibroblasts (CAFs) revealed heterogeneity in the TME of PAC. Intercellular communication analysis showed strong direct interactions between matrix CAFs, inflammatory CAFs, and epithelial cells. Additionally, we found that the SPP1-associated pathway was activated in monocytes, whereas the vascular endothelial growth factor-associated pathway was activated in epithelial cells. These results improve the understanding of the TME of pancreatic cancer and provide a foundation for further studies on intratumoral heterogeneity. In addition, differentially expressed gene secretory leukocyte protease inhibitor (SLPI) was identified in pancreatic cancer, and functional experiments showed that SLPI had a strong impact on cell viability and apoptosis, which offers a potential therapy target for pancreatic cancer.
Collapse
Affiliation(s)
- Bolun Ai
- The Faculty of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yicheng Liang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Yan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yangyang Lei
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Gou S, Liu N, Liu W, Yao Y. Knowledge relay: Synergetic generation and transfer learning for pancreatic tumor segmentation on multimodal images. Med Phys 2025; 52:4828-4843. [PMID: 40032630 DOI: 10.1002/mp.17713] [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: 05/14/2024] [Revised: 01/28/2025] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Pancreatic cancer is among the most lethal malignancies, with the lowest survival rates. The use of image-guided radiotherapy has shown significant potential in enhancing surgical outcomes for pancreatic cancer. However, accurate segmentation of pancreatic tumors prior to radiotherapy remains a challenge due to the small size, irregular shape, and indistinct boundaries of the pancreas and tumor in monomodal imaging. Furthermore, the availability of multimodal images that meet the requirements for precise pancreatic segmentation is highly limited, leading to the datasets that fail to provide comprehensive knowledge for effective image representation in pancreatic tumor segmentation. PURPOSE This study aims to develop a method for accurately segmenting pancreatic tumors under very harsh data conditions, in which the currently available datasets are fragmented with the issues, such as limited sample sizes, inconsistent lesion matching, and incomplete modalities. METHODS We propose a knowledge relay framework that leverages synergistic generation and transfer learning strategies. The relay comprises three batons: pancreatic PET image generation, coarse detection, and fine segmentation. Multimodal images, including CT, MR, and PET from three separate datasets, are integrated within this framework. The knowledge contained in each dataset is sequentially transferred and aggregated through the batons by the strategies of transfer learning and fine-tuning. Additionally, we introduce a mask-constrained CycleGAN and an inter-attention UNet within this framework to enhance the extraction and utilization of knowledge for accurate pancreatic tumor segmentation. RESULTS The proposed knowledge relay framework achieves the state-of-the-art performance in pancreatic tumor segmentation on PET/MR images. On the images collected from 19 subjects, our method attained a DSC $\text{DSC}$ of 80.06%, SEN $\text{SEN}$ of 83.39%, SPE $\text{SPE}$ of 99.81%, ASD $\text{ASD}$ of 4.87 mm $\text{mm}$ , and95 HD $95 \text{HD}$ of 12.69 mm $\text{mm}$ . CONCLUSIONS The results of comparison and ablation experiments validate the effectiveness of the proposed knowledge relay framework in extracting and integrating knowledge from fragmented datasets under constrained conditions. The comprehensive and enriched knowledge significantly enhances the accuracy of pancreatic tumor segmentation.
Collapse
Affiliation(s)
- Shuiping Gou
- Key Laboratory of Intelligent Perception and Image Understanding of Ministry of Education, School of Artificial Intelligence, Xidian University, Xi'an, Shannxi, China
| | - Ningtao Liu
- Key Laboratory of Intelligent Perception and Image Understanding of Ministry of Education, School of Artificial Intelligence, Xidian University, Xi'an, Shannxi, China
| | - Wenbo Liu
- Translational Medicine Research Center of the First Affiliated Hospital, Weifang Medical University, Weifang, Shandong, China
| | - Yao Yao
- School of Information Engineering, Hangzhou Vocational and Technical College, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Chuong MD, Ashman J, Jethwa K, Kharofa J, Kim H, Koay E, Ludmir E, Miller E, Nelson B, Reyngold M, Sanford N, Chang D. Moving From the Background Toward the Spotlight: A Critical Review of Radiation Therapy for Locally Advanced Pancreas Cancer. Int J Radiat Oncol Biol Phys 2025; 122:294-312. [PMID: 40032056 DOI: 10.1016/j.ijrobp.2025.02.022] [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: 08/30/2024] [Revised: 01/31/2025] [Accepted: 02/16/2025] [Indexed: 03/05/2025]
Abstract
Radiation therapy (RT) for locally advanced pancreatic cancer (LAPC) continues to be controversial. Advances in both systemic therapy and RT techniques have changed the landscape of LAPC management in recent years. Clinical outcomes of ablative RT have been encouraging, and randomized clinical trials may clarify the role of RT for LAPC. We present a contemporary critical review of key aspects regarding optimal patient selection, radiation dose escalation techniques, novel radiosensitizers and radioprotectors, and treatment response assessment for LAPC.
Collapse
Affiliation(s)
- Michael D Chuong
- Department of Radiation Oncology, Miami Cancer Institute, Miami, Florida.
| | - Jonathan Ashman
- Department of Radiation Oncology, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Krishan Jethwa
- Department of Radiation Oncology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Jordan Kharofa
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Hyun Kim
- Department of Radiation Oncology, Washington University in St. Louis, Missouri.
| | - Eugene Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ethan Ludmir
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eric Miller
- Department of Radiation Oncology, Ohio State University, Columbus, Ohio
| | - Bailey Nelson
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Marsha Reyngold
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nina Sanford
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Daniel Chang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
6
|
Fu Y, Chen L, Lv N, Wang J, Yu S, Fang Q, Xin W. miR-135b-5p/PDE3B Axis Regulates Gemcitabine Resistance in Pancreatic Cancer Through Epithelial-Mesenchymal Transition. Mol Carcinog 2025; 64:1119-1130. [PMID: 40170518 DOI: 10.1002/mc.23914] [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: 11/19/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/03/2025]
Abstract
Gemcitabine-based chemotherapy is an effective treatment for pancreatic cancer (PC), but gemcitabine resistance frequently compromises the therapeutic efficacy, resulting in clinical chemotherapeutic failure and a poor prognosis for patients. In this study, we investigated the mechanisms of gemcitabine chemoresistance in PC by examining the roles of microRNAs linked to gemcitabine resistance and their downstream signaling pathways. In vitro experiments were performed to alter miR-135b-5p levels in PC parental and drug-resistant cells to probe its function. miR-135b-5p targets PDE3B was confirmed by using RNA-seq technology to screen for gemcitabine-resistance-associated mRNAs in PC. A series of rescue experiments were performed after cotransfection, demonstrating that PDE3B could reverse miR-135b-5p-mediated chemoresistance and epithelial-mesenchymal transition (EMT). These findings indicate that the miR-135b-5p/PDE3B axis generates resistance by stimulating the EMT signaling pathway, which provides new insights into gemcitabine chemoresistance in PC.
Collapse
Affiliation(s)
- Yuxuan Fu
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Liangsheng Chen
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Neng Lv
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Jia Wang
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Shuwei Yu
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Qilu Fang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Wenxiu Xin
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Selvam SP, Kamalakannan S, Mathivanan A, Cho S. Engineering Pt Single-Atom Doped SeS 2/Ti 3CNT x MXene with Molecularly Imprinted Polymer for Precision Pancreatic Cancer Diagnostics: DFT and Molecular Dynamics Perspectives. SMALL METHODS 2025:e2500475. [PMID: 40420649 DOI: 10.1002/smtd.202500475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 05/11/2025] [Indexed: 05/28/2025]
Abstract
Cyclophilin-B (CypB) is overexpressed in pancreatic cancer, thus, the potential screening of CypB in biofluids and tissue samples may boost the identification of early-stage pancreatic cancer. A novel strategy of CypB detection utilizing the molecularly imprinted polymer platform, comprising higher binding affinity exhibiting cavities against the CypB protein was developed. Specifically, a nanocatalyst consisting of Pt single atom (Ptsa)-doped selenium disulfide (SeS2)/Ti3CNTx MXene nanocomposite is designed. The sluggish diffusion of Ptsa caused by the highest migration energy barrier of 6.39 eV unveils exceptionally high stability (2.89 ×1088 d (300 K) and 1.053 × 1024 d (750 K)) with (SeS2)/Ti3CNTx surface. The Ptsa boosted charge transfer kinetics paves the improved performance of the CypB sensor, while SeS2/Ti3CNTx supports the stable current density overall. The system establishes a dynamic linear range from 0.12 to 250 nm of CypB detection which correlates with the physiological existence of the CypB in human biofluids and tissues and the excellent detection limit of 80 pm. The liquid chromatography integrated mass spectrometer investigation warranted the significant enhancement of CypB associates with the progression of pancreatic cancer.
Collapse
Affiliation(s)
- Sathish Panneer Selvam
- Department of Electronic Engineering, Gachon University, Seongnam-si, Gyeonggi-do, 13120, South Korea
| | | | - Agalya Mathivanan
- Department of Physics, Sri Manakula Vinayagar Engineering College, Madagadipet, Puducherry, 605107, India
| | - Sungbo Cho
- Department of Electronic Engineering, Gachon University, Seongnam-si, Gyeonggi-do, 13120, South Korea
- Department of Health Sciences and Technology, Gachon University, Incheon, 21999, South Korea
- Department of Semiconductor Engineering, Gachon University, Seongnam-si, Gyeonggi-do, 13120, South Korea
| |
Collapse
|
8
|
Hüttner FJ, Klotz R, Giese NA, Kong B, Ahmed A, Merz D, Pöchmann A, Burghaus I, Hackert T, Strobel O, Mihaljevic AL, Michalski CW, Büchler MW, Diener MK. Pancreatic resection with perioperative drug repurposing of propranolol and etodolac - the phase II randomized controlled PROSPER trial. Langenbecks Arch Surg 2025; 410:168. [PMID: 40402347 PMCID: PMC12098435 DOI: 10.1007/s00423-025-03735-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2025] [Accepted: 05/07/2025] [Indexed: 05/23/2025]
Abstract
PURPOSE The perioperative period is characterized by psychological stress and inflammatory reactions that can contribute to disease recurrence or metastatic spread. These reactions are mediated particularly by catecholamines and prostaglandins. The PROSPER trial aimed to evaluate whether a perioperative drug repurposing with a non-selective betablocker (propranolol) and a COX-2 inhibitor (etodolac) is feasible and safe in the setting of pancreatic cancer surgery. METHODS Patients undergoing partial pancreatoduodenectomy for pancreatic cancer were randomized to perioperative treatment with propranolol and etodolac or placebo. Main safety endpoint was the rate of serious adverse events (SAE) and the main feasibility endpoint was adherence. Overall and disease-free survival (DFS) as well as recurrences were assessed as efficacy parameters and the trial was accompanied by a translational study. RESULTS The trial was prematurely closed due to slow recruitment. 26 patients were randomized, but 6 never started trial medication. Finally, 9 patients received the trial medication and 11 patients placebo. There were 6 SAE in the treatment vs. 14 in the placebo group. Adherence was lower in the treatment group, but without statistically significance. Median DFS was 16.36 months (95%-CI 1.18 - not reached) in verum vs. 11.25 (95%-CI 2.2 - 17.25) in placebo group. The rate of distant recurrences was 11.1% in verum vs. 54.5% in placebo group. CONCLUSION There were no safety concerns, but the trial intervention was not feasible given slow recruitment and limited adherence. However, the translational study and preliminary efficacy data revealed some promising findings, warranting further investigation. REGISTRATION DRKS00014054.
Collapse
Affiliation(s)
- Felix J Hüttner
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Department of General, Visceral and Thoracic Surgery, Klinikum Nürnberg, Prof.-Ernst-Nathan-Str. 1, 90419, Nuremberg, Germany
| | - Rosa Klotz
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Study Center of the German Surgical Society (SDGC), Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Nathalia A Giese
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Bo Kong
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Azaz Ahmed
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Department of Cancer Immunology and Cancer Immunotherapy, German Cancer Research Center, Heidelberg, Germany
| | - Daniela Merz
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Alexandra Pöchmann
- Department of Cancer Immunology and Cancer Immunotherapy, German Cancer Research Center, Heidelberg, Germany
- Helmholtz Institute for Translational Oncology (HI-TRON), Mainz, Germany
| | - Ina Burghaus
- Coordination Centre for Clinical Trials (KKS), Medical Faculty & Heidelberg University Hospital, Heidelberg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver Strobel
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - André L Mihaljevic
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Department of General, Visceral and Transplantation Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Christoph W Michalski
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Markus W Büchler
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Botton-Champalimaud Pancreatic Cancer, Champalimaud Foundation, Lisbon, Portugal
| | - Markus K Diener
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany.
- Department of General, Visceral and Thoracic Surgery, Klinikum Nürnberg, Prof.-Ernst-Nathan-Str. 1, 90419, Nuremberg, Germany.
| |
Collapse
|
9
|
Wang T, Song W, Tang Y, Yi J, Pan H. Breaking the immune desert: Strategies for overcoming the immunological challenges of pancreatic cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189353. [PMID: 40412630 DOI: 10.1016/j.bbcan.2025.189353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 05/18/2025] [Accepted: 05/18/2025] [Indexed: 05/27/2025]
Abstract
Pancreatic cancer is characterised by its highly aggressive nature and extremely poor prognosis, with a uniquely complex tumour immune microenvironment that manifests as a prototypical "immune desert." This immune-desert phenotype primarily arises from the inherently low immunogenicity of the tumour, the formation of a dense fibrotic stroma, severe deficiency in immune cell infiltration, and profound immunosuppressive effects of the metabolic landscape. Specifically, dysregulated tryptophan metabolism, such as indoleamine 2,3-dioxygenase (IDO)-mediated catabolism, and excessive lactate accumulation contribute to impaired T-cell functionality. Collectively, these factors severely limit the efficacy of current immunotherapy strategies, particularly those based on immune checkpoint inhibitors, which have demonstrated significantly lower clinical response rates in pancreatic cancer than in other malignancies. In response to these therapeutic challenges, this review explores integrated treatment strategies that combine metabolic reprogramming, tumour microenvironment remodelling, and next-generation immune checkpoint blockades, such as LAG-3, TIM-3, and VISTA. These emerging approaches hold substantial promise for clinical application. For example, targeting key metabolic pathways, including glycolysis (Warburg effect) and glutamine metabolism, may help restore T-cell activity by alleviating metabolic stress within the tumour milieu. Additionally, localised administration of immune stimulators such as interleukin-12 (IL-12) and CD40 agonists may enhance immune cell infiltration and promote tumour-specific immune activation. Future research should prioritise large-scale, multicentre clinical trials to validate the therapeutic efficacy of these innovative strategies, aiming to achieve meaningful breakthroughs in pancreatic cancer immunotherapy and significantly improve long-term survival and clinical outcomes in affected patients.
Collapse
Affiliation(s)
- Tianming Wang
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China; Zhengning County Community Health Service Center, Qingyang 745300, Gansu Province, China
| | - Wenjing Song
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Yuan Tang
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Jianfeng Yi
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China.
| | - Haibang Pan
- First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China.
| |
Collapse
|
10
|
Qi J, Fang C, Deng C, Shi F, Yao Q. Mesoporous Magnetic Graphene for Serum Metabolic Profiling in Non-Invasive Early Detection and Diagnosis of Pancreatic Ductal Adenocarcinoma. ACS APPLIED MATERIALS & INTERFACES 2025; 17:29995-30005. [PMID: 40355809 DOI: 10.1021/acsami.5c03176] [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: 05/15/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive and lethal cancer, typically diagnosed at advanced stages due to its asymptomatic onset and challenges in early detection. To address the critical need for the early diagnosis of PDAC, we developed a laser desorption/ionization mass spectrometry (LDI-MS) platform based on mesoporous silica-modified magnetic graphene (MG@mSiO2). MG@mSiO2 exhibited exceptional ultraviolet (UV) absorption, efficient ionization, and minimal background interference, enabling high-resolution profiling of serum metabolic fingerprints (SMFs). Based on the extracted SMFs, we constructed a Random Forest (RF) model to classify PDAC patients, high-risk (HR) individuals, and healthy controls (HC), achieving an accuracy of 97.5% in the independent test set. Additionally, a six-metabolite biomarker panel was identified, showing strong diagnostic potential with sensitivity and accuracy exceeding 89.1% for distinguishing HC from PDAC. When coupled with the serological marker carbohydrate antigen 19-9 (CA19-9), the integrated strategy delivered significantly improved diagnostic performance, achieving high accuracy ranging from 95.3% to 100% in distinguishing HR and PDAC patients from HC. Furthermore, metabolic pathway analysis revealed key pathways associated with PDAC progression, providing mechanistic insights into the disease. This work provides a powerful diagnostic tool for PDAC screening, establishing a foundation for early detection and precision medicine in clinical practice.
Collapse
Affiliation(s)
- Jia Qi
- Department of Chemistry, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Caiyun Fang
- Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Chunhui Deng
- Department of Chemistry, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Fangying Shi
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China
| | - Qunyan Yao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Gastroenterology and Hepatology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, China
| |
Collapse
|
11
|
Dubrova A, Cavaniol C, Van de Walle A, Mathieu P, Fusilier Z, Yaacoub N, Lalatonne Y, Descroix S, Wilhelm C. Magnetite Nanoparticle Photothermal Therapy in a Pancreatic Tumor-on-Chip: A Dual-Action Approach Targeting Cancer Cells and their Microenvironment. ACS NANO 2025. [PMID: 40397413 DOI: 10.1021/acsnano.5c02099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
The application of magnetite nanoparticles (MagNPs) for photothermal therapy (MagNP-PTT) has recently expanded to cancer treatment. This study introduces MagNP-PTT in a tumor-on-a-chip model to target highly aggressive pancreatic ductal adenocarcinoma (PDAC). A tumor-on-chip system was developed using PANC-1 PDAC cells embedded in a collagen type I extracellular matrix and cultured for 1 week to form tumor spheroids. This platform offers a framework for applying PTT in a model system that aims to mimic the native tumor microenvironment. MagNPs efficiently penetrate the tumor spheroids, achieving controlled heating via near-infrared (NIR) light. By adjusting nanoparticle concentration and laser power, temperature increments of 2 °C between 38-48 °C were established. Temperatures above 44 °C significantly increased cell death, while lower temperatures allowed partial recovery. Beyond inducing cancer cell death, MagNP-PTT altered the extracellular matrix and triggered a slight epithelial-mesenchymal transition marked by increased vimentin expression. These findings highlight MagNP-PTT as a dual-action therapy, targeting both tumor cells and their microenvironment, offering an alternative approach for overcoming stromal barriers in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Anastasiia Dubrova
- Laboratoire Physique des Cellules et Cancer, PCC, CNRS UMR168, Institut Curie, Sorbonne Université, PSL Research University, 75005 Paris, France
| | - Charles Cavaniol
- Laboratoire Physique des Cellules et Cancer, PCC, CNRS UMR168, Institut Curie, Sorbonne Université, PSL Research University, 75005 Paris, France
| | - Aurore Van de Walle
- Laboratoire Physique des Cellules et Cancer, PCC, CNRS UMR168, Institut Curie, Sorbonne Université, PSL Research University, 75005 Paris, France
| | - Paul Mathieu
- Université Sorbonne Paris Nord, Université Paris Cité, Laboratory for Vascular Translational Science, LVTS, INSERM, UMR 1148, Bobigny F-93017, France
| | - Zoé Fusilier
- Institut Curie, PSL University, INSERM U932, Immunity and Cancer, 75005 Paris, France
| | - Nader Yaacoub
- Institut des Molécules et Materiaux du Mans, CNRS UMR-6283, Le Mans Université, F-72085 Le Mans, France
| | - Yoann Lalatonne
- Université Sorbonne Paris Nord, Université Paris Cité, Laboratory for Vascular Translational Science, LVTS, INSERM, UMR 1148, Bobigny F-93017, France
- Département de Biophysique et de Médecine Nucléaire, Assistance Publique-Hôpitaux de Paris, Hôpital Avicenne F- 93009, Bobigny, France
| | - Stephanie Descroix
- Laboratoire Physique des Cellules et Cancer, PCC, CNRS UMR168, Institut Curie, Sorbonne Université, PSL Research University, 75005 Paris, France
| | - Claire Wilhelm
- Laboratoire Physique des Cellules et Cancer, PCC, CNRS UMR168, Institut Curie, Sorbonne Université, PSL Research University, 75005 Paris, France
| |
Collapse
|
12
|
Guan X, Mu Y, Jin X, Wang C. Integrated multi-omics profiling to establish an IGFBP-based prognostic score for pancreatic ductal adenocarcinoma: unraveling prognostic biomarkers, immune microenvironment crosstalk, and therapeutic implications. Front Immunol 2025; 16:1600527. [PMID: 40443651 PMCID: PMC12119506 DOI: 10.3389/fimmu.2025.1600527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Accepted: 04/24/2025] [Indexed: 06/02/2025] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is accompanied by endocrine dysfunction, particularly involving dysregulation of the insulin and insulin-like growth factor (IGF) signaling pathways. Clinical manifestations such as hyperglycemia and insulin resistance are common and have been linked to aberrant expression of insulin-like growth factor-binding proteins (IGFBPs). However, the specific roles and mechanisms of IGFBP family genes in PDAC remain unclear. Method We conducted a multi-dimensional integrative analysis using publicly available PDAC cohorts, stratifying patients based on IGFBP gene expression profiles. A prognostic model was constructed to classify patients into risk groups. To explore the biological mechanisms underlying IGFBP involvement in PDAC, we further incorporated single-cell transcriptomic sequencing and spatial transcriptomic data to investigate the relationship between IGFBP expression and the tumor immune microenvironment. Result Our prognostic model effectively stratified PDAC patients into distinct risk categories with significant survival differences. High-risk patients demonstrated specific IGFBP expression patterns associated with aggressive tumor biology. Single-cell and spatial transcriptomic analyses revealed that IGFBP family genes modulate immune cell infiltration and spatial immune heterogeneity within the tumor microenvironment. Conclusion This study identified the IGFBP family genes as key modulators of PDAC progression and immune landscape remodeling. These findings supported the potential of IGFBP family genes as prognostic biomarkers and therapeutic targets, offering new insights into PDAC biology and opportunities for personalized treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Chengfeng Wang
- State Key Lab of Molecular Oncology and Department of Pancreatic and Gastric Surgery,
National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
13
|
Roscigno G, Jacobs S, Toledo B, Borea R, Russo G, Pepe F, Serrano MJ, Calabrò V, Troncone G, Giovannoni R, Giovannetti E, Malapelle U. The potential application of stroma modulation in targeting tumor cells: focus on pancreatic cancer and breast cancer models. Semin Cancer Biol 2025:S1044-579X(25)00060-4. [PMID: 40373890 DOI: 10.1016/j.semcancer.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer development and spreading being considered as "the dark side of the tumor". Within this term tumor cells, immune components, supporting cells, extracellular matrix and a myriad of bioactive molecules that synergistically promote tumor development and therapeutic resistance, are included. Recent findings revealed the profound impacts of TME on cancer development, serving as physical support, critical mediator and biodynamic matrix in cancer evolution, immune modulation, and treatment outcomes. TME targeting strategies built on vasculature, immune checkpoints, and immuno-cell therapies, have paved the way for revolutionary clinical interventions. On this basis, the relevance of pre-clinical and clinical investigations has rapidly become fundamental for implementing novel therapeutical strategies breaking cell-cell and cell -mediators' interactions between TME components and tumor cells. This review summarizes the key players in the breast and pancreatic TME, elucidating the intricate interactions among cancer cells and their essential role for cancer progression and therapeutic resistance. Different tumors such breast and pancreatic cancer have both different and similar stroma features, that might affect therapeutic strategies. Therefore, this review aims to comprehensively evaluate recent findings for refining breast and pancreatic cancer therapies and improve patient prognoses by exploiting the TME's complexity in the next future.
Collapse
Affiliation(s)
- Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy.
| | - Sacha Jacobs
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| | - Belen Toledo
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén E-23071, Spain.
| | - Roberto Borea
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| | - Gianluca Russo
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Maria Jose Serrano
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy; GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception Group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain.
| | - Viola Calabrò
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Roberto Giovannoni
- Department of Biology, Genetic Unit, University of Pisa, Via Derna 1, 56126 Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy.
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy.
| |
Collapse
|
14
|
Liu PJ, Zhou ZP, Wang GY, Xu S, Wang W, Chen X, Tan XD, Liu ZH, Zhao ZM, Gao YX, Zhang XP, Liu R. New-onset diabetes worsens prognosis of patients with pancreatic ductal adenocarcinoma after R0 resection: A multicenter study. Hepatobiliary Pancreat Dis Int 2025:S1499-3872(25)00088-8. [PMID: 40374469 DOI: 10.1016/j.hbpd.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 04/03/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND The risk of pancreatic ductal adenocarcinoma (PDAC) is increased in patients with diabetes mellitus (DM), particularly in new-onset diabetes (NOD). This study aimed to analyze the effect of NOD on the outcomes of patients with PDAC after R0 resection. METHODS PDAC patients from six centers in China undergoing R0 resection from 2015 to 2022 were included. Patients were categorized as long-term diabetes (LTD), NOD, or non-diabetes mellitus (non-DM) based on the timing of diagnosis relative to pancreatic resection. We compared the effects of diabetes status on perioperative and oncological outcomes of PDAC. RESULTS Of 1211 patients, 602 (49.7%), 127 (10.5%), and 482 (39.8%) were in the non-DM, LTD, and NOD groups, respectively. Patients with NOD suffered from higher rates of fatty pancreas and postoperative pancreatic fistula (POPF) (both P < 0.05). When compared with the non-DM group, the NOD group had worse median overall survival (OS) (24.6 vs. 29.4 months, P < 0.001) and recurrence-free survival (RFS) (13.3 vs. 15.8 months, P < 0.001); and the LTD group also had worse median OS (25.2 vs. 29.4 months, P = 0.041) and RFS (13.8 vs. 15.8 months, P = 0.007) compared with non-DM group. However, there were no significant differences in survival between the NOD and the LTD groups. Multivariate analysis indicated that NOD, LTD, largest tumor size, and poor tumor differentiation were independently associated with worse OS and RFS (all P < 0.05). CONCLUSIONS Patients with PDAC undergoing R0 resection experienced a higher probability of POPF in the presence of concurrent NOD. Long-term survival prognosis was worse in NOD or LTD patients than in non-DM patients.
Collapse
Affiliation(s)
- Peng-Jiong Liu
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Zhi-Peng Zhou
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Guan-Yu Wang
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Shuai Xu
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Wei Wang
- Department of General Surgery, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Xiong Chen
- Department of Hepatobiliary Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, China
| | - Xiao-Dong Tan
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Zhong-Hua Liu
- Department of Hepatobiliary Surgery, Chifeng Municipal Hospital, Chifeng 024050, China
| | - Zhi-Ming Zhao
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Yuan-Xing Gao
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Xiu-Ping Zhang
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| | - Rong Liu
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
15
|
Yamane K, Anazawa T, Nagai K, Kasai Y, Masui T, Izuwa A, Kurahashi K, Ishida S, Ogiso S, Yoshimura M, Iwai T, Matsubara J, Fukuda A, Isoda H, Hidaka Y, Ibi Y, Hatano E. Neoadjuvant Chemoradiotherapy Using Moderately Hypofractionated Intensity-Modulated Radiotherapy Versus Upfront Surgery for Resectable Pancreatic Cancer: A Retrospective Cohort Study. Ann Surg Oncol 2025; 32:3603-3613. [PMID: 39893341 PMCID: PMC11976822 DOI: 10.1245/s10434-025-16956-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/16/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND The efficacy of neoadjuvant chemoradiotherapy for resectable pancreatic ductal adenocarcinoma (R-PDAC) remains unclear. This study was designed to evaluate neoadjuvant chemoradiotherapy by using intensity-modulated radiotherapy (NAC-IMRT) for R-PDAC compared with upfront surgery (UpS). METHODS Among 198 patients with R-PDAC who were indicated for resection between 2013 and 2021, 130 were included in this study after excluding patients who underwent neoadjuvant chemotherapy and did not meet the NAC-IMRT criteria (Eligible set). NAC-IMRT was planned for 58 patients, and UpS was planned for 72 patients. Additionally, in 105 patients who could undergo the planned treatment (As-treated set), the surgical, pathological, and oncological outcomes were evaluated. RESULTS In the Eligible set, median overall survival (OS) was 50.5 months with NAC-IMRT and 34.7 months with UpS and progression-free survival was 20.4 months with NAC-IMRT and 13.9 months with UpS. In the As-treated set, OS was longer in the NAC-IMRT group (66.7 months vs. 34.7 months, p = 0.007). On multivariate analysis, NAC-IMRT was identified as an independent factor for better OS (hazard ratio 0.617, 95% confidence interval 0.382-0.995, p = 0.047, in the Eligible set). The incidence of postoperative complications did not show a difference between the two groups, and NAC-IMRT suppressed local tumor invasion, including lymphatic, venous, perineural invasion, and lymph node metastases. CONCLUSIONS NAC-IMRT may offer superior survival outcomes and manageable toxicity in R-PDAC patients compared with upfront surgery. This study supports the efficacy and safety of NAC-IMRT and recommends its consideration in R-PDAC treatment protocols.
Collapse
Affiliation(s)
- Kei Yamane
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takayuki Anazawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Kazuyuki Nagai
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yosuke Kasai
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshihiko Masui
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Surgery, Kurashiki Central Hospital, Okayama, Japan
| | - Aya Izuwa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koki Kurahashi
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Ishida
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Ogiso
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Michio Yoshimura
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiro Iwai
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junichi Matsubara
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyoshi Isoda
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yu Hidaka
- Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yumiko Ibi
- Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Etsuro Hatano
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
16
|
Tufo A, Milanetto AC, Valente R, Spalice E, Sodano L, Pasquali C, Scandavini MC, Coppola A. The role of indocyanine green in fluorescence-guided pancreatic surgery: a comprehensive review. Int J Surg 2025; 111:3386-3398. [PMID: 40009558 PMCID: PMC12165567 DOI: 10.1097/js9.0000000000002311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 02/11/2025] [Indexed: 02/28/2025]
Abstract
Pancreatic surgery is a complex and challenging field, with patients facing a high risk of postoperative complications. In recent years, indocyanine green (ICG) has gained prominence as a valuable tool used in various aspects of pancreatic surgery. ICG is a fluorescent dye that offers real-time imaging capabilities that enhance the surgeon's ability to accurately localize tumors and critical anatomical structures, thereby improving surgical precision and potentially reducing operative time and complications. One of the most significant advantages of ICG is its ability to provide enhanced visualization of the biliary tract and vascular structures, which is particularly beneficial in complex pancreatic resections, in which the anatomy can be highly variable and challenging to navigate. Furthermore, ICG can be instrumental in ensuring the adequate perfusion of anastomoses, thereby reducing the risk of postoperative leaks and associated morbidity. This comprehensive review aims to provide an in-depth analysis of the current applications, advantages, and limitations of ICG in pancreatic surgery.
Collapse
Affiliation(s)
- Andrea Tufo
- UOC Chirurgia Generale, Ospedale del Mare, Napoli, Italy
| | | | - Roberto Valente
- Department of Diagnostic and Intervention, Surgery, Umeå University, Umeå, Sweden
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Enrico Spalice
- Department of Surgery, Sapienza University of Rome, Rome, Italy
| | | | - Claudio Pasquali
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | | | | |
Collapse
|
17
|
Wang Y, Guo J, Chen M, Liao S, Xu L, Chen Q, Song G, Zhang XB. Ultrabright and ultrafast afterglow imaging in vivo via nanoparticles made of trianthracene derivatives. Nat Biomed Eng 2025; 9:656-670. [PMID: 39472533 DOI: 10.1038/s41551-024-01274-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 09/16/2024] [Indexed: 05/22/2025]
Abstract
Low sensitivity, photobleaching, high-power excitation and long acquisition times constrain the utility of afterglow luminescence. Here we report the design and imaging performance of nanoparticles made of electron-rich trianthracene derivatives that, on excitation by room light at ultralow power (58 μW cm-2), emit afterglow luminescence at ~500 times those of commonly used organic afterglow nanoparticles. The nanoparticles' ultrabright afterglow allowed for deep-tissue imaging (up to 6 cm), for ultrafast afterglow imaging (at short acquisition times down to 0.01 s) of naturally behaving mice with negligible photobleaching, even after re-excitation for over 15 cycles, and for the accurate visualization of subcutaneous and orthotopic tumours and of plaque in carotid arteries. We also show that an afterglow nanoparticle that is activated only in the presence of granzyme B allowed for the tracking of granzyme-B activity in the context of therapeutic monitoring. The high sensitivity and negligible photobleaching of the organic afterglow nanoparticles offer advantages for real-time in vivo monitoring of physiopathological processes.
Collapse
Affiliation(s)
- Youjuan Wang
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Jing Guo
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Muchao Chen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Shiyi Liao
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Li Xu
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Qian Chen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, China
| | - Guosheng Song
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China.
| | - Xiao-Bing Zhang
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China.
| |
Collapse
|
18
|
Zou L, Cao Y, Nie Z, Mao L, Qiu Y, Wang Z, Cai Z, Yang X. Segment Like A Doctor: Learning reliable clinical thinking and experience for pancreas and pancreatic cancer segmentation. Med Image Anal 2025; 102:103539. [PMID: 40112510 DOI: 10.1016/j.media.2025.103539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/05/2025] [Accepted: 02/27/2025] [Indexed: 03/22/2025]
Abstract
Pancreatic cancer is a lethal invasive tumor with one of the worst prognosis. Accurate and reliable segmentation for pancreas and pancreatic cancer on computerized tomography (CT) images is vital in clinical diagnosis and treatment. Although certain deep learning-based techniques have been tentatively applied to this task, current performance of pancreatic cancer segmentation is far from meeting the clinical needs due to the tiny size, irregular shape and extremely uncertain boundary of the cancer. Besides, most of the existing studies are established on the black-box models which only learn the annotation distribution instead of the logical thinking and diagnostic experience of high-level medical experts, the latter is more credible and interpretable. To alleviate the above issues, we propose a novel Segment-Like-A-Doctor (SLAD) framework to learn the reliable clinical thinking and experience for pancreas and pancreatic cancer segmentation on CT images. Specifically, SLAD aims to simulate the essential logical thinking and experience of doctors in the progressive diagnostic stages of pancreatic cancer: organ, lesion and boundary stage. Firstly, in the organ stage, an Anatomy-aware Masked AutoEncoder (AMAE) is introduced to model the doctors' overall cognition for the anatomical distribution of abdominal organs on CT images by self-supervised pretraining. Secondly, in the lesion stage, a Causality-driven Graph Reasoning Module (CGRM) is designed to learn the global judgment of doctors for lesion detection by exploring topological feature difference between the causal lesion and the non-causal organ. Finally, in the boundary stage, a Diffusion-based Discrepancy Calibration Module (DDCM) is developed to fit the refined understanding of doctors for uncertain boundary of pancreatic cancer by inferring the ambiguous segmentation discrepancy based on the trustworthy lesion core. Experimental results on three independent datasets demonstrate that our approach boosts pancreatic cancer segmentation accuracy by 4%-9% compared with the state-of-the-art methods. Additionally, the tumor-vascular involvement analysis is also conducted to verify the superiority of our method in clinical applications. Our source codes will be publicly available at https://github.com/ZouLiwen-1999/SLAD.
Collapse
Affiliation(s)
- Liwen Zou
- School of Mathematics, Nanjing University, Nanjing, 210093, China
| | - Yingying Cao
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210000, China
| | - Ziwei Nie
- School of Mathematics, Nanjing University, Nanjing, 210093, China
| | - Liang Mao
- Department of Pancreatic Surgery, Nanjing Drum Tower Hospital, Nanjing, 210008, China
| | - Yudong Qiu
- Department of Pancreatic Surgery, Nanjing Drum Tower Hospital, Nanjing, 210008, China
| | - Zhongqiu Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210000, China
| | - Zhenghua Cai
- Department of Pancreatic Surgery, Nanjing Drum Tower Hospital, Nanjing, 210008, China; Medical School, Nanjing University, Nanjing, 210007, China.
| | - Xiaoping Yang
- School of Mathematics, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
19
|
Calheiros J, Silva R, Barbosa F, Morais J, Moura SR, Almeida S, Fiorini E, Mulhovo S, Aguiar TQ, Wang T, Ricardo S, Almeida MI, Domingues L, Melo SA, Corbo V, Ferreira MJU, Saraiva L. A first-in-class inhibitor of homologous recombination DNA repair counteracts tumour growth, metastasis and therapeutic resistance in pancreatic cancer. J Exp Clin Cancer Res 2025; 44:129. [PMID: 40275348 PMCID: PMC12020112 DOI: 10.1186/s13046-025-03389-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is among the cancer types with poorest prognosis and survival rates primarily due to resistance to standard-of-care therapies, including gemcitabine (GEM) and olaparib. Particularly, wild-type (wt)BRCA tumours, the most prevalent in PDAC, are more resistant to DNA-targeting agents like olaparib, restraining their clinical application. Recently, we disclosed a monoterpene indole alkaloid derivative (BBIT20) as a new inhibitor of homologous recombination (HR) DNA repair with anticancer activity in breast and ovarian cancer. Since inhibition of DNA repair enhances the sensitivity of cancer cells to chemotherapy, we aimed to investigate the anticancer potential of BBIT20 against PDAC, particularly carrying wtBRCA. METHODS In vitro and in vivo PDAC models, particularly human cell lines (including GEM-resistant PDAC cells), patient-derived organoids and xenograft mice of PDAC were used to evaluate the anticancer potential of BBIT20, alone and in combination with GEM or olaparib. Disruption of the BRCA1-BARD1 interaction by BBIT20 was assessed by co-immunoprecipitation, immunofluorescence and yeast two-hybrid assay. RESULTS The potent antiproliferative activity of BBIT20, superior to olaparib, was demonstrated in PDAC cells regardless of BRCA status, by inducing cell cycle arrest, apoptosis, and DNA damage, while downregulating HR. The disruption of DNA double-strand breaks repair by BBIT20 was further reinforced by non-homologous end joining (NHEJ) suppression. The inhibition of BRCA1-BARD1 heterodimer by BBIT20 was demonstrated in PDAC cells and confirmed in a yeast two-hybrid assay. In GEM-resistant PDAC cells, BBIT20 showed potent antiproliferative, anti-migratory and anti-invasive activity, overcoming GEM resistance by inhibiting the multidrug resistance P-glycoprotein, upregulating the intracellular GEM-transporter ENT1, and downregulating GEM resistance-related microRNA-20a and GEM metabolism enzymes as RRM1/2. Furthermore, BBIT20 did not induce resistance in PDAC cells. It inhibited the growth of patient-derived PDAC organoids, by inducing apoptosis, repressing HR, and potentiating olaparib and GEM cytotoxicity. The enhancement of olaparib antitumor activity by BBIT20 was confirmed in xenograft mice of PDAC. Notably, it hindered tumour growth and liver metastasis formation, improving survival of orthotopic xenograft mice of PDAC. Furthermore, its potential as a stroma-targeting agent, reducing fibrotic extracellular matrix and overcoming desmoplasia, associated with an enhancement of immune cell response by depleting PD-L1 expression in tumour tissues, renders BBIT20 even more appealing for combination therapy, particularly with immunotherapy. CONCLUSION These findings underscore the great potential of BBIT20 as a novel multifaceted anticancer drug candidate for PDAC treatment.
Collapse
Grants
- 2020.04613.BD FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2020.06020.BD FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- 2022.05718.PTDC, 0.54499/LA/P/0008/2020, 10.54499/UIDP/50006/2020, 10.54499/UIDB/50006/2020 FCT/MCTES, Fundação para a Ciência e Tecnologia and Ministério da Ciência, Tecnologia e Ensino Superior
- AIRC; IG No 288801 Associazione Italiana Ricerca sul Cancro
- AIRC; IG No 288801 Associazione Italiana Ricerca sul Cancro
- NHI; HHSN26100008 NCI NIH HHS
- National Cancer Institute
Collapse
Affiliation(s)
- Juliana Calheiros
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Rita Silva
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Filipa Barbosa
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa, 1649-003, Portugal
| | - João Morais
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Sara Reis Moura
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Sofia Almeida
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Elena Fiorini
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
| | - Silva Mulhovo
- Centro de Estudos Moçambicanos e de Etnociências (CEMEC), Faculty of Natural Sciences and Mathematics, Pedagogical University, Maputo, 21402161, Mozambique
| | - Tatiana Q Aguiar
- CEB - Centre of Biological Engineering, University of Minho, Braga, 4710-057, Portugal
- LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| | - Tao Wang
- Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, China
| | - Sara Ricardo
- Associate Laboratory i4HB - Institute for Health and Bioeconomy and UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra, 4585-116, Portugal
| | - Maria Inês Almeida
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
| | - Lucília Domingues
- CEB - Centre of Biological Engineering, University of Minho, Braga, 4710-057, Portugal
- LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| | - Sonia A Melo
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal
- Department of Pathology, Faculty of Medicine University of Porto, Al. Prof. Hernâni Monteiro, Porto, 4200-319, Portugal
- Porto Comprehensive Cancer Centre (P.CCC) Raquel Seruca, Porto, Portugal
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
| | - Maria-José U Ferreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa, 1649-003, Portugal.
| | - Lucília Saraiva
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal.
| |
Collapse
|
20
|
Wang N, Yan X, Ji C, Hu J, Chen X, Zhang C, Quan Y, He T, Sun T, Yu Y. In situ thermal-responsive hydrogels for combined photothermal therapy and chemotherapy of pancreatic cancer. RSC Adv 2025; 15:13119-13126. [PMID: 40275869 PMCID: PMC12019213 DOI: 10.1039/d5ra00371g] [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: 01/15/2025] [Accepted: 02/14/2025] [Indexed: 04/26/2025] Open
Abstract
Pancreatic cancer is a malignancy with a poor prognosis and high mortality. Survival outcomes remain very poor despite significant advances in molecular diagnostics and therapeutics in clinical practice. Surgical resection is the only potentially curative treatment, but the tumor is often diagnosed at an advanced stage, and most cancers recur after surgery. Treatments other than surgery, including chemotherapy and immunotherapy, still offer disappointing results. Multidisciplinary treatment approaches through appropriate carriers have provided new solutions for improving the prognosis of pancreatic cancer. Herein, we reported an in situ formed thermo-responsive hybrid hydrogel loaded with gemcitabine and manganese dioxide nanoparticles, which exhibited good injectability, high photothermal hyperthermia, and biocompatibility, leading to efficient multidisciplinary treatment of pancreatic cancer in combination with chemotherapy and photothermal therapy (PTT). The hybrid hydrogel could be heated to 51 °C under 808 nm laser irradiation in five minutes. In situ intratumoral injection results suggested that the hybrid hydrogel exhibited high photothermal efficiency in killing rabbit pancreatic tumors. In vivo results indicated that the multidisciplinary treatment almost completely eliminated subcutaneous tumors in mice within 14 days. This development offers an efficient multidisciplinary treatment for pancreatic cancer.
Collapse
Affiliation(s)
- Ningwei Wang
- Department of Gastroenterology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China Hefei 230001 China
| | - Xu Yan
- Anhui Province Key Laboratory of Value-Added Catalytic Conversion and Reaction Engineering, Anhui Province Engineering Research Center of Flexible and Intelligent Materials, School of Chemistry and Chemical Engineering, Hefei University of Technology Hefei 230001 China
| | - Chaofei Ji
- Department of Radiology, Anhui No. 2 Provincial People's Hospital Hefei 230001 China
| | - Jinlong Hu
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital Hefei 230001 China
| | - Xiangshun Chen
- Anhui Province Key Laboratory of Value-Added Catalytic Conversion and Reaction Engineering, Anhui Province Engineering Research Center of Flexible and Intelligent Materials, School of Chemistry and Chemical Engineering, Hefei University of Technology Hefei 230001 China
| | - Cong Zhang
- Department of Gastroenterology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China Hefei 230001 China
| | - Yuzhu Quan
- Changchun GeneScience Pharmaceutical Co., Ltd. Xuhui District Shanghai 200030 China
| | - Tao He
- Anhui Province Key Laboratory of Value-Added Catalytic Conversion and Reaction Engineering, Anhui Province Engineering Research Center of Flexible and Intelligent Materials, School of Chemistry and Chemical Engineering, Hefei University of Technology Hefei 230001 China
| | - Tianci Sun
- Anhui Province Key Laboratory of Value-Added Catalytic Conversion and Reaction Engineering, Anhui Province Engineering Research Center of Flexible and Intelligent Materials, School of Chemistry and Chemical Engineering, Hefei University of Technology Hefei 230001 China
| | - Yue Yu
- Department of Gastroenterology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China Hefei 230001 China
| |
Collapse
|
21
|
An P, Wang J, Fan R. Identifying and validating PLAU as a potential prognostic biomarker for PDAC. Sci Rep 2025; 15:12515. [PMID: 40216916 PMCID: PMC11992124 DOI: 10.1038/s41598-025-97629-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a prevalent cancer with a high mortality rate. This study aims to identify and validate biomarkers for early PDAC diagnosis. We employed the GEO2R online tool to screen differentially expressed genes (DEGs), construct protein interaction networks, and perform functional enrichment analysis, survival prognosis analysis, and expression level validation. We identified 260 DEGs, comprising 165 upregulated genes and 95 downregulated genes. Following functional enrichment and survival analysis, we selected plasminogen activator urokinase (PLAU) for the RNA and protein level verification and preliminary cell phenotype analysis. We found that PLAU knockdown inhibits the proliferation and survival of pancreatic cancer cells. Therefore, PLAU may serve as a potential biomarker, offering new strategies for understanding PDAC's pathological mechanisms.
Collapse
Affiliation(s)
- Peng An
- Material Evidence Technology Center, School of Law, Tianjin University of Commerce, Tianjin, 300134, China.
| | - Junhong Wang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Rong Fan
- Central Laboratory, Tianjin Xiqing hospital, Tianjin, 300380, PR China.
| |
Collapse
|
22
|
Hasan R, Zhao Z, Li Y, Liu Y, Zhang Y, Cheng K. Small extracellular vesicles (sEVs) in pancreatic cancer progression and diagnosis. J Control Release 2025; 380:269-282. [PMID: 39889882 PMCID: PMC11908897 DOI: 10.1016/j.jconrel.2025.01.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
Pancreatic cancer is one of the most aggressive malignancies with poor prognostic outcomes, necessitating the exploration of novel biomarkers and therapeutic targets for early detection and effective treatment. Small extracellular vesicles (sEVs) secreted by cells, have gained considerable attention in cancer research due to their role in intercellular communication and their potential as non-invasive biomarkers. This review focuses on the role of sEVs in the progression of pancreatic cancer and their application as biomarkers. We delve into the biogenesis, composition, and functional implications of sEVs in pancreatic tumor biology, emphasizing their involvement in processes such as tumor growth, metastasis, immune modulation, and chemotherapy resistance. In addition, we discuss the challenges in isolating and characterizing sEVs. The review also highlights recent advances in the utilization of sEV-derived biomarkers for the early diagnosis, prognosis, and monitoring of pancreatic cancer. By synthesizing the latest findings, we aim to underscore the significance of sEVs in pancreatic cancer and their potential to revolutionize patient management through improved diagnostics and targeted therapies.
Collapse
Affiliation(s)
- Reaid Hasan
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Zhen Zhao
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Yuanke Li
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Yanli Liu
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Kun Cheng
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA.
| |
Collapse
|
23
|
Takano Y, Tamai N, Yamawaki M, Noda J, Azami T, Niiya F, Nishimoto F, Maruoka N, Yamagami T, Nagahama M. The outcomes of endoscopic ultrasound-guided tissue acquisition for small focal liver lesions measuring ≤2 cm. DEN OPEN 2025; 5:e70031. [PMID: 39439546 PMCID: PMC11494021 DOI: 10.1002/deo2.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/27/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
Objectives Endoscopic ultrasound-guided tissue acquisition (EUS-TA) for focal liver lesions has gained attention as an alternative to percutaneous biopsy. Although the outcomes of EUS-TA for focal liver lesions have been reported to be favorable, no studies have focused on small focal liver lesions (≤2 cm). The aim of this study was to evaluate the outcomes of EUS-TA for small focal liver lesions (≤2 cm). Methods The details of EUS-TA performed for focal liver lesions between 2016 and 2022 were retrospectively reviewed. The outcomes were compared between cases involving ≤2 cm lesions and those involving >2 cm lesions. The primary outcomes were diagnostic ability and adverse events. Results EUS-TA for focal liver lesions was performed in 109 cases. Of the 109 cases, 32 (29.3%) involved ≤2 cm lesions and 77 (70.6%) involved >2 cm lesions. Right lobe lesions and transduodenal puncture were significantly fewer in the ≤2 cm group. There were no significant differences in needle gauge, needle type, or number of punctures between the groups. The sensitivity, specificity, and accuracy rates were 96.8%, 100%, and 96.8%, respectively, in the ≤2 cm group and 97.4%, 100%, and 97.4%, respectively, in the >2 cm group, with no significant differences between the groups. There was no difference in adverse events between the groups (0% in the ≤2 cm group and 2.3% in the >2 cm group). Conclusions EUS-TA for small focal liver lesions measuring ≤2 cm has favorable outcomes, which are similar to those for lesions measuring >2 cm.
Collapse
Affiliation(s)
- Yuichi Takano
- Department of Internal MedicineDivision of Gastroenterology, Showa University Fujigaoka HospitalKanagawaJapan
| | - Naoki Tamai
- Department of Internal MedicineDivision of Gastroenterology, Showa University Fujigaoka HospitalKanagawaJapan
| | - Masataka Yamawaki
- Department of Internal MedicineDivision of Gastroenterology, Showa University Fujigaoka HospitalKanagawaJapan
| | - Jun Noda
- Department of Internal MedicineDivision of Gastroenterology, Showa University Fujigaoka HospitalKanagawaJapan
| | - Tetsushi Azami
- Department of Internal MedicineDivision of Gastroenterology, Showa University Fujigaoka HospitalKanagawaJapan
| | - Fumitaka Niiya
- Department of Internal MedicineDivision of Gastroenterology, Showa University Fujigaoka HospitalKanagawaJapan
| | - Fumiya Nishimoto
- Department of Internal MedicineDivision of Gastroenterology, Showa University Fujigaoka HospitalKanagawaJapan
| | - Naotaka Maruoka
- Department of Internal MedicineDivision of Gastroenterology, Showa University Fujigaoka HospitalKanagawaJapan
| | - Tatsuya Yamagami
- Department of Internal MedicineDivision of Gastroenterology, Showa University Fujigaoka HospitalKanagawaJapan
| | - Masatsugu Nagahama
- Department of Internal MedicineDivision of Gastroenterology, Showa University Fujigaoka HospitalKanagawaJapan
| |
Collapse
|
24
|
Sastre J, Pérez S, Sabater L, Rius-Pérez S. Redox signaling in the pancreas in health and disease. Physiol Rev 2025; 105:593-650. [PMID: 39324871 DOI: 10.1152/physrev.00044.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024] Open
Abstract
This review addresses oxidative stress and redox signaling in the pancreas under healthy physiological conditions as well as in acute pancreatitis, chronic pancreatitis, pancreatic cancer, and diabetes. Physiological redox homeodynamics is maintained mainly by NRF2/KEAP1, NF-κB, protein tyrosine phosphatases, peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α), and normal autophagy. Depletion of reduced glutathione (GSH) in the pancreas is a hallmark of acute pancreatitis and is initially accompanied by disulfide stress, which is characterized by protein cysteinylation without increased glutathione oxidation. A cross talk between oxidative stress, MAPKs, and NF-κB amplifies the inflammatory cascade, with PP2A and PGC1α as key redox regulatory nodes. In acute pancreatitis, nitration of cystathionine-β synthase causes blockade of the transsulfuration pathway leading to increased homocysteine levels, whereas p53 triggers necroptosis in the pancreas through downregulation of sulfiredoxin, PGC1α, and peroxiredoxin 3. Chronic pancreatitis exhibits oxidative distress mediated by NADPH oxidase 1 and/or CYP2E1, which promotes cell death, fibrosis, and inflammation. Oxidative stress cooperates with mutant KRAS to initiate and promote pancreatic adenocarcinoma. Mutant KRAS increases mitochondrial reactive oxygen species (ROS), which trigger acinar-to-ductal metaplasia and progression to pancreatic intraepithelial neoplasia (PanIN). ROS are maintained at a sufficient level to promote cell proliferation, while avoiding cell death or senescence through formation of NADPH and GSH and activation of NRF2, HIF-1/2α, and CREB. Redox signaling also plays a fundamental role in differentiation, proliferation, and insulin secretion of β-cells. However, ROS overproduction promotes β-cell dysfunction and apoptosis in type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Salvador Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Luis Sabater
- Liver, Biliary and Pancreatic Unit, Hospital Clínico, Department of Surgery, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Sergio Rius-Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
- Department of Cell Biology, Functional Biology and Physical Anthropology, Faculty of Biology, University of Valencia, Valencia, Spain
| |
Collapse
|
25
|
Xu W, Hu L, Shi S, Gao J, Ye J, Lu Y. Prediction of Potential Drugs Targeting Acute Pancreatitis Based on the HLA-DR-Related Gene-Monocyte Infiltration Regulatory Network. Biomed Eng Comput Biol 2025; 16:11795972251328458. [PMID: 40165943 PMCID: PMC11956513 DOI: 10.1177/11795972251328458] [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: 10/02/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Background Acute pancreatitis (AP) is a common disease of acute abdominal pain, the incidence of which is increasing annually, but its pathogenesis remains incompletely understood. Methods Gene expression profiles of AP were obtained from the Gene Expression Omnibus (GEO) database. R software was used to identify differentially expressed genes (DEGs) and perform functional analysis. The diagnostic value of HLA-DR-related genes was assessed by receiver operating characteristic (ROC) curves. Monocyte infiltration abundance in AP and normal groups was analyzed by Cibersort method, and the correlation between HLA-DR-related genes and monocyte abundance was analyzed. The modules highly correlated with HLA-DR-related genes were clarified by WGCNA modeling, and the core genes regulating HLA-DR were obtained by using LASSO regression. Finally, potential drugs targeting the above genes were analyzed by Enrichr database. Result A Total of 3 HLA-DR-related genes (HLA-DRA, HLA-DRB1, and HLA-DRB5) were identified, which were negatively correlated with the severity of AP and had excellent disease diagnostic value (AUC = 0.761, 0.761, and 0.718), were were positively correlated with monocyte abundance. We identified 110 genes that positively regulate HLA-DR and 130 genes that negatively regulate HLA-DR. LASSO regression identified UCP2, GK, and SAMHD1 as the core nodes of the regulated genes. Compared with the normal group, UCP2 and SAMHD1 were reduced in AP, and the opposite was true for GK, and SAMHD1 had better sensitivity and specificity in diagnosing AP. Drug sensitivity analysis predicted 12 drugs acting on HLA-DRA, HLA-DRB1, and HLA-DRB5 and 8 drugs acting on UCP2, GK, and SAMHD1. Conclusion We identified 3 HLA-DR-related genes (HLA-DRA, HLA-DRB1, and HLA-DRB5) and 3 coregulatory nodes (UCP2, GK, and SAMHD1), which were associated with AP severity and monocyte abundance. Based on these genes, we predicted 20 potential therapeutic agents for AP.
Collapse
Affiliation(s)
- Wei Xu
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lan Hu
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengyi Shi
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Gao
- Division of Critical Care, Nanxiang Hospital of Jiading District, Shanghai, China
| | - Jing Ye
- Department of Geriatrics, Medical Center on Aging, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine; International Laboratory in Hematology and Cancer, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d’Azur University, Shanghai, China
- The State Key Laboratory of Medical Genomics, Pôle Sino-Français de Recherche en Sciences Du Vivant et Génomique, Shanghai, China
| | - Yiming Lu
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Division of Critical Care, Nanxiang Hospital of Jiading District, Shanghai, China
- The State Key Laboratory of Medical Genomics, Pôle Sino-Français de Recherche en Sciences Du Vivant et Génomique, Shanghai, China
| |
Collapse
|
26
|
Cui R, Wang G, Hu R, Wang Y, Mu H, Song Y, Chen B, Jiang X. Prognostic and immunotherapeutic potential of disulfidptosis-associated signature in pancreatic cancer. Front Immunol 2025; 16:1568976. [PMID: 40207217 PMCID: PMC11979277 DOI: 10.3389/fimmu.2025.1568976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Disulfidptosis is a newly discovered formation of programmed cell death. However, the significance of disulfidptosis in pancreatic adenocarcinoma remains unclear. Our investigation aims to elucidate the significance of disulfidptosis in pancreatic ductal adenocarcinoma by integrating diverse datasets, including bulk RNA sequencing data, microarray profiles, single-cell transcriptome profiles, spatial transcriptome data, and biospecimens. Utilizing various bioinformatics tools, we screened disulfidptosis-related genes based on single-cell RNA sequencing profiles, subsequently validating them through enrichment analysis. An 8-gene disulfidptosis-related prognostic signature was established by constructing massive LASSO-Cox regression models and validated by multiple external PDAC cohorts. Evaluation methods, such as Kaplan-Meier curves, ROC curves, time-dependent ROC curves, and decision curve analysis, were employed to assess the prognostic signature's reliability. High disulfidptosis-related scores were associated with a poorer prognosis and diminished sensitivity to immune checkpoint blockade. Further investigation uncovered that the potential components of elevated DPS involve malignant tumor hallmarks, extensive interactions between myCAFs and tumor cells, and the exclusion of immune cells. Cell-cell communication analysis highlighted myCAFs' role in signaling, potentially influencing tumor cells towards increased malignancy through collagen, laminin, and FN1 signaling networks. Spatial transcriptome analysis confirmed the crosstalk between myCAFs and tumor cells. Biospecimens including 20 pairs of PDAC samples and adjacent normal tissues further demonstrated the robustness of DPS and its correlation with CAF markers. In conclusion, our study introduces a novel disulfidptosis-related signature with high efficacy in patient risk stratification, which has the ability to predict the sensitivity to immune checkpoint blockade.
Collapse
Affiliation(s)
- Ran Cui
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Gaoming Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Renhao Hu
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yongkun Wang
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huiling Mu
- Department of Biobank, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanxiang Song
- Department of Biobank, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bo Chen
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaohua Jiang
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
27
|
Meng Y, Wang C, Usyk M, Kwak S, Peng C, Hu KS, Oberstein PE, Krogsgaard M, Li H, Hayes RB, Ahn J. Association of tumor microbiome with survival in resected early-stage PDAC. mSystems 2025; 10:e0122924. [PMID: 40013793 PMCID: PMC11915875 DOI: 10.1128/msystems.01229-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
The pancreas tumor microbiota may influence tumor microenvironment and influence survival in early-stage pancreatic ductal adenocarcinoma (PDAC); however, current studies are limited and small. We investigated the relationship of tumor microbiota to survival in 201 surgically resected patients with localized PDAC (Stages I-II), from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) cohorts. We characterized the tumor microbiome using RNA-sequencing data. We examined the association of the tumor microbiome with overall survival (OS), via meta-analysis with the Cox PH model. A microbial risk score (MRS) was calculated from the OS-associated microbiota. We further explored whether the OS-associated microbiota is related to host tumor immune infiltration. PDAC tumor microbiome α- and β-diversities were not associated with OS; however, 11 bacterial species, including species of Gammaproteobacteria, confirmed by extensive resampling, were significantly associated with OS (all Q < 0.05). The MRS summarizing these bacteria was related to a threefold change in OS (hazard ratio = 2.96 per standard deviation change in the MRS, 95% confidence interval = 2.26-3.86). This result was consistent across the two cohorts and in stratified analyses by adjuvant therapy (chemotherapy/radiation). Identified microbiota and the MRS also exhibited association with memory B cells and naïve CD4+ T cells, which may be related to the immune landscape through BCR and TCR signaling pathways. Our study shows that a unique tumor microbiome structure, potentially affecting the tumor immune microenvironment, is associated with poorer survival in resected early-stage PDAC. These findings suggest that microbial mechanisms may be involved in PDAC survival, potentially informing PDAC prognosis and guiding personalized treatment strategies.IMPORTANCEMuch of the available data on the PDAC tumor microbiome and survival are derived from relatively small and heterogeneous studies, including those involving patients with advanced stages of pancreatic cancer. There is a critical knowledge gap in terms of the tumor microbiome and survival in early-stage patients treated by surgical resection; we expect that advancements in survival may initially be best achieved in these patients who are treated with curative intent.
Collapse
Affiliation(s)
- Yixuan Meng
- Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
| | - Chan Wang
- Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
| | - Mykhaylo Usyk
- Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
| | - Soyoung Kwak
- Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
| | - Chengwei Peng
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Kenneth S. Hu
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Paul E. Oberstein
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
| | - Michelle Krogsgaard
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
| | - Huilin Li
- Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
| | - Richard B. Hayes
- Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
| | - Jiyoung Ahn
- Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
- NYU Laura and Isaac Perlmutter Cancer Center, New York, New York, USA
| |
Collapse
|
28
|
Wang Z, Xie X, Xue Y, Chen Y. Tryptophan-2,3-Dioxygenase as a Therapeutic Target in Digestive System Diseases. BIOLOGY 2025; 14:295. [PMID: 40136551 PMCID: PMC11939885 DOI: 10.3390/biology14030295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025]
Abstract
Tryptophan (Trp) is an essential amino acid that must be acquired exclusively through dietary intake. The metabolism of tryptophan plays a critical role in maintaining immune homeostasis and tolerance, as well as in preventing excessive inflammatory responses. Tryptophan-2,3-dioxygenase (TDO2) is a tetrameric heme protein and serves as one of the pivotal rate-limiting enzymes in the first step of tryptophan metabolism. Dysregulation of TDO2 expression has been observed in various digestive system diseases, encompassing those related to the oral cavity, esophagus, liver, stomach, pancreas, and colon and rectum. Digestive system diseases are the most common clinical diseases, with complex clinical manifestations and interrelated symptoms, and have become a research hotspot in the field of medicine. Studies have demonstrated that aberrant TDO2 expression is closely associated with various clinical manifestations and disease outcomes in patients with digestive system disorders. Consequently, TDO2 has garnered increasing recognition as a promising therapeutic target for digestive system diseases in recent years, attracting growing attention. This article provides a brief overview of the role of TDO2 in the tryptophan pathway, emphasizing its significant involvement in diseases of the digestive system. Strategies targeting TDO2 through specific inhibitors suggest considerable promise in enhancing therapeutic outcomes for digestive diseases. Thus, this review concludes by discussing recent advancements in the development of TDO2 inhibitors. We believe that targeted inhibition of TDO2 combined with immunotherapy, the screening of a large number of natural products, and the assistance of artificial intelligence in drug design will be important directions for developing more effective TDO2 inhibitors and improving treatment outcomes in the future.
Collapse
Affiliation(s)
| | | | | | - Yixuan Chen
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China
| |
Collapse
|
29
|
Grobbelaar C, Steenkamp V, Mabeta P. Vascular Endothelial Growth Factor Receptors in the Vascularization of Pancreatic Tumors: Implications for Prognosis and Therapy. Curr Issues Mol Biol 2025; 47:179. [PMID: 40136433 PMCID: PMC11941243 DOI: 10.3390/cimb47030179] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 03/27/2025] Open
Abstract
In pancreatic cancer (PC), vascular endothelial growth factor (VEGF) and its primary receptor, vascular endothelial growth factor receptor (VEGFR)-2, are central drivers of angiogenesis and metastasis, with their overexpression strongly associated with poor prognosis. In some PC patients, VEGF levels correlate with disease stage, tumor burden, and survival outcomes. However, therapies targeting VEGF and VEGFR-2, including tyrosine kinase inhibitors (TKIs) and monoclonal antibodies, have demonstrated limited efficacy, partly due to the emergence of resistance mechanisms. Resistance appears to stem from the activation of alternative vascularization pathways. This review explores the multifaceted roles of VEGFRs in pancreatic cancer, including VEGFR-1 and VEGFR-3. Potential strategies to improve VEGFR-targeting therapies, such as combination treatments, the development of more selective inhibitors, and the use of biomarkers, are discussed as promising approaches to enhance treatment efficacy and outcomes.
Collapse
Affiliation(s)
- Craig Grobbelaar
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0002, South Africa;
| | - Vanessa Steenkamp
- Department of Pharmacology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0002, South Africa;
| | - Peace Mabeta
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0002, South Africa;
| |
Collapse
|
30
|
Wu HY, Tsou HH, Lu LS, Lee HL, Chiou JF, Ch'ang HJ. Role of Neoadjuvant Chemoradiation Therapy for Resectable and Borderline Resectable Pancreatic Adenocarcinoma-A Systematic Review and Meta-Analysis. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00175-0. [PMID: 40074045 DOI: 10.1016/j.ijrobp.2025.02.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/31/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Randomized trials and meta-analyses have indicated longer survival with neoadjuvant than with adjuvant therapy in patients with resectable or borderline resectable (R/BR) pancreatic adenocarcinoma. Despite the efficacy of chemotherapy, the role of radiation therapy as an adjuvant or neoadjuvant treatment for patients with R/BR pancreatic adenocarcinoma remains unclear. In this systematic review and meta-analysis, we compared the benefits of additional chemoradiation therapy (CRT) to neoadjuvant chemotherapy (NAC) with NAC alone for R/BR pancreatic adenocarcinoma. METHODS AND MATERIALS A systematic literature search was conducted on Embase, Web of Science, PubMed, Cochrane, and Google Scholar. Median overall survival (OS) was the primary endpoint. Secondary endpoints included disease-free survival (DFS), resection rate, and R0 resection rate. RESULTS This review and meta-analysis included 31 prospective studies, of which 9 were randomized trials. In these studies, 658 patients from 14 study arms received NAC alone and 912 patients from 19 study arms received both NAC and CRT (NAC-CRT). The pooled median OS was 25.55 months (95% CI, 21.59-30.24 months) for NAC alone and 17.55 months (95% CI, 16.47-18.70 months; P < .0001) for NAC-CRT. The pooled R0 resection rate was higher with NAC-CRT (83.43%) than with NAC (69.97%; P < .0001). No significant difference was observed in DFS or resection rate between the 2 groups. In patients who received 5 or more cycles of initial chemotherapy, NAC-CRT was associated with longer OS than NAC (23.30 vs 21.85 months; P = .856). CONCLUSIONS NAC provides significantly longer OS than NAC-CRT to R/BR pancreatic adenocarcinoma. NAC-CRT is associated with a significantly improved R0 resection rate. This positive local effect of CRT can be translated to extended survival when 5 cycles or more of NAC are prescribed.
Collapse
Affiliation(s)
- Hsiao-Yu Wu
- Institute of Public Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Hsiao-Hui Tsou
- Institute of Public Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Long-Sheng Lu
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hsin-Lun Lee
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Jeng Fong Chiou
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hui-Ju Ch'ang
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan; Department of Oncology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
31
|
Ko CC, Yang PM. Hypoxia-induced MIR31HG expression promotes partial EMT and basal-like phenotype in pancreatic ductal adenocarcinoma based on data mining and experimental analyses. J Transl Med 2025; 23:305. [PMID: 40065368 PMCID: PMC11895263 DOI: 10.1186/s12967-025-06292-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is the most common and aggressive type of pancreatic cancer, with a five-year survival rate below 8%. Its high mortality is largely due to late diagnosis, metastatic potential, and resistance to therapy. Epithelial-mesenchymal transition (EMT) plays a key role in metastasis, enabling cancer cells to become mobile. Partial EMT, where cells maintain both epithelial and mesenchymal traits, is more frequent in tumors than complete EMT and contributes to cancer progression. The long non-coding RNA MIR31 host gene (MIR31HG) has recently emerged as a critical factor in PDAC oncogenesis. This study aimed to investigate MIR31HG's role in partial EMT and its association with the basal-like PDAC subtype. METHODS We analyzed the relationship between MIR31HG expression, partial EMT, and the basal-like subtype of PDAC by integrating data from public databases. We reanalyzed public data from PDAC patient-derived organoids to assess MIR31HG expression and gene signatures under hypoxic and normoxic conditions. RNA sequencing and bioinformatics analyses, including gene set enrichment analysis (GSEA), were used to investigate differentially expressed genes and pathway enrichments. EMT, partial EMT, and hypoxia scores were calculated based on the expression levels of specific gene sets. RESULTS We observed that MIR31HG overexpression strongly correlates with higher partial EMT scores and the stabilization of the epithelial phenotype in PDAC. MIR31HG is highly expressed in the basal-like subtype of PDAC, which exhibits partial EMT traits. Hypoxia, a hallmark of basal-like PDAC, was shown to significantly induce MIR31HG expression, thereby promoting the basal-like phenotype and partial EMT. In patient-derived organoids, hypoxic conditions increased MIR31HG expression and enhanced basal-like and partial EMT gene signatures, while normoxia reduced these expressions. These findings suggest that hypoxia-induced MIR31HG expression plays a crucial role in driving the aggressive basal-like subtype of PDAC. CONCLUSIONS Our results indicate that MIR31HG is crucial in regulating PDAC progression, particularly in the aggressive basal-like subtype associated with hypoxia and partial EMT. Targeting the MIR31HG-mediated network may offer a novel therapeutic approach to combat hypoxia-driven PDAC.
Collapse
Affiliation(s)
- Ching-Chung Ko
- Department of Medical Imaging, Chi Mei Medical Center, Tainan, 71004, Taiwan
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, 71710, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Pei-Ming Yang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, No. 301, Yuantong Rd., Zhonghe Dist., New Taipei City, 235603, Taiwan.
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei, 11031, Taiwan.
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei, 11696, Taiwan.
- Taipei Cancer Center, Taipei Medical University (TMU) and Affiliated Hospitals Pancreatic Cancer Groups, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
32
|
Phulara NR, Ishida CT, Espenshade PJ, Seneviratne HK. Gemcitabine Alters Phosphatidylcholine Metabolism in Mouse Pancreatic Tumors. J Proteome Res 2025; 24:1209-1218. [PMID: 39973059 DOI: 10.1021/acs.jproteome.4c00839] [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] [Indexed: 02/21/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest diseases, despite advancements in elucidating tumor biology and developing novel therapeutics. Importantly, lipids, such as phospholipids, are crucial for the survival and proliferation of tumor cells. However, the impact of chemotherapeutic drugs on phospholipid metabolism in PDAC remains poorly understood. Gemcitabine (a nucleoside analogue) is a first-line drug in PDAC treatment, but its clinical effectiveness is limited by multiple factors. Herein, we employed matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) and proteomics approaches to investigate gemcitabine-induced lipid metabolism alterations in mouse pancreatic tumors following gemcitabine treatment (n = 3, control tumors; n = 3, gemcitabine-treated tumors). From MALDI MSI experiments, we observed elevated levels of several phosphatidylcholines (PCs), PC(30:0), PC(32:3), PC(34:2), PC(36:1), and PC(36:2), in gemcitabine-treated tumor tissues compared to the control. In addition, proteomics data revealed the differential abundance of several phospholipid-binding proteins in response to gemcitabine treatments. Furthermore, several endoplasmic reticulum stress-related proteins exhibited high expression in gemcitabine-treated tumor tissues. Altogether, our MALDI MSI and proteomics data provide important insights into alterations in PC metabolism in pancreatic tumors in response to gemcitabine treatment. Importantly, targeting the altered PC metabolism during gemcitabine therapy might help combat pancreatic cancer.
Collapse
Affiliation(s)
- Nav Raj Phulara
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| | - Chiaki Tsuge Ishida
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Peter John Espenshade
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Giovanis Institute for Translational Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Herana Kamal Seneviratne
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| |
Collapse
|
33
|
Tokuyama N, Ikeda S, Ishida R, Futai R, Tobimatsu K, Kodama Y. Impaired Consciousness Due to Hyperammonemia During S-1 Administration for Unresectable Pancreatic Cancer. Intern Med 2025; 64:695-698. [PMID: 39048368 PMCID: PMC11949670 DOI: 10.2169/internalmedicine.3771-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/28/2024] [Indexed: 07/27/2024] Open
Abstract
A 71-year-old woman diagnosed with unresectable locally advanced pancreatic cancer was initially treated with gemcitabine and nab-paclitaxel as first-line therapy. The tumor exhibited no significant progression; however, after 12 cycles, the patient developed drug-induced interstitial pneumonia, leading to the discontinuation of gemcitabine and nab-paclitaxel therapy. Following recovery from pneumonia, S-1 therapy was initiated as second-line treatment. During S-1 therapy, she was hospitalized because of impaired consciousness and was subsequently diagnosed with hyperammonemia induced by S-1. Although rarely reported, S-1-induced hyperammonemia is potentially a significant adverse effect. Here, we herein report the case of a patient with pancreatic cancer.
Collapse
Affiliation(s)
- Nagahiro Tokuyama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Japan
| | - Sayaka Ikeda
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Japan
| | - Ryosuke Ishida
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Japan
| | - Ryoko Futai
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Japan
| | - Kazutoshi Tobimatsu
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Japan
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Japan
| |
Collapse
|
34
|
Malarz K, Korzuch J, Mrozek-Wilczkiewicz A, Szubka M, Rurka P, Małota K, Herraiz A, Dreszer D, Kocot K, Herranz F, Rost-Roszkowska M, Sun T, Musioł R, Serda M. Aminofullerenes as targeted inhibitors of EGFR: from pancreatic cancer inhibitors to Drosophila m. Toxicology. Nanomedicine (Lond) 2025; 20:585-601. [PMID: 39916650 PMCID: PMC11881853 DOI: 10.1080/17435889.2025.2461985] [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: 08/28/2024] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
AIM Pancreatic ductal adenocarcinoma (PDAC) is recognized as one of the most formidable cancers, largely due to its distinct microenvironment characterized predominantly by extensive desmoplastic stroma. In this study, we synthesized three novel water-soluble fullerene-based nanomaterials targeting EGFR protein. METHODS The direct amination of fullerene carbon atoms, was followed by conjugation with a modified derivative of the EGFR inhibitor-erlotinib, resulting in the formation of novel water-soluble fullerene derivatives. RESULTS Further investigation into PAN02 and AsPC-1 cell lines revealed that these fullerene nanomaterials could induce cell cycle arrest in the G0/G1 phase, corroborated by alterations in the expression levels of the p27 and cyclin E1 proteins. Additionally, mechanisms of cell death were identified as autophagy for C60BUT and C70BUT-ERL, and apoptosis for Gd@C82EDA-ERL nanomaterials. CONCLUSIONS Crucially, the study uncovered the efficacy of synthesized aminofullerenes in inhibiting the EGFR signaling pathway. The further toxicological studies of Gd@C82EDA-ERL fullerene on Drosophila melanogaster, underscored its potential for theranostic applications.
Collapse
Affiliation(s)
- Katarzyna Malarz
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
- Institute of Physics, Faculty of Science and Technology, University of Silesia in Katowice, Chorzów, Poland
| | - Julia Korzuch
- Institute of Chemistry, Faculty of Science and Technology, University of Silesia in Katowice, Katowice, Poland
| | - Anna Mrozek-Wilczkiewicz
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
- Institute of Physics, Faculty of Science and Technology, University of Silesia in Katowice, Chorzów, Poland
| | - Magdalena Szubka
- Institute of Physics, Faculty of Science and Technology, University of Silesia in Katowice, Chorzów, Poland
| | - Patryk Rurka
- Institute of Physics, Faculty of Science and Technology, University of Silesia in Katowice, Chorzów, Poland
| | - Karol Małota
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia in Katowice, Katowice, Poland
| | - Aitor Herraiz
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Dominik Dreszer
- Institute of Chemistry, Faculty of Science and Technology, University of Silesia in Katowice, Katowice, Poland
| | - Karina Kocot
- Institute of Chemistry, Faculty of Science and Technology, University of Silesia in Katowice, Katowice, Poland
| | - Fernando Herranz
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Magdalena Rost-Roszkowska
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia in Katowice, Katowice, Poland
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Robert Musioł
- Institute of Chemistry, Faculty of Science and Technology, University of Silesia in Katowice, Katowice, Poland
| | - Maciej Serda
- Institute of Chemistry, Faculty of Science and Technology, University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
35
|
Azizzadeh B, Majidinia M, Gheysarzadeh A. The reciprocal effects of autophagy and the Warburg effect in pancreatic ductal adenocarcinoma: an in vitro study. Med Oncol 2025; 42:86. [PMID: 40021508 DOI: 10.1007/s12032-025-02631-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/13/2025] [Indexed: 03/03/2025]
Abstract
Autophagy and the Warburg effect are two common pathways in pancreatic ductal adenocarcinoma (PDAC). To date, the reciprocal effects of these pathways have not yet been elucidated. Therefore, this study was designed to investigate the relationship between these factors in vitro and may provide therapeutic targets in the future. The Mia-Paca-2 and AsPc-1 cell lines were cultured under normal conditions. To achieve autophagy, starvation was induced by Hank's balanced salt solution (HBSS), whereas autophagy was inhibited by 3-methyladenine (3-MA). The Warburg effect is mimicked by lactic acid, and the Warburg effect is inhibited by oxamate, the main inhibitor of lactate dehydrogenase. Cell viability was checked through the MTT assay method. Autophagy was checked via evaluation of Beclin-1 via western blotting. The amount of lactic acid was also measured with a lactate dehydrogenase (LDH) assay kit. The cells were incubated with different concentrations of 3-MA, lactic acid and oxamate. The viability of AsPc-1 cells decreased, and the IC50 values were 1195 µM, 23.06 mM and 8.617 mM for 3-MA, lactic acid and oxamate, respectively. Similarly, the IC50 values of Mia-Paca-2 were 873.9 µM, 35.9 mM and 26.74 mM for 3-MA, lactic acid and oxamate, respectively. Our data revealed that starvation increased the expression of the autophagy-related protein Beclin-1 (P value < 0.05); however, 3-MA significantly reduced its expression (P value < 0.05). In addition, lactic acid alone did not affect the expression level of Beclin-1 (P value > 0.05), but oxamate treatment increased its expression (P value < 0.05). We also showed that starvation reduced lactic acid levels, but an autophagy inhibitor, 3MA, significantly increased lactic acid production (P value < 0.05). Our findings showed that lactic acid alone has no significant effect on autophagy and that oxamate induces autophagy, possibly because of caloric restriction. On the other hand, autophagy inhibits lactic acid production, whereas the inhibition of autophagy leads to increased lactic acid production.
Collapse
Affiliation(s)
- Bita Azizzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Gheysarzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran.
| |
Collapse
|
36
|
Hu ZY, Ding D, Song Y, Deng YF, Zhang CM, Yu T. Molecular mechanism of pancreatic ductal adenocarcinoma: The heterogeneity of cancer-associated fibroblasts and key signaling pathways. World J Clin Oncol 2025; 16:97007. [PMID: 39995552 PMCID: PMC11686552 DOI: 10.5306/wjco.v16.i2.97007] [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: 05/20/2024] [Revised: 10/04/2024] [Accepted: 11/04/2024] [Indexed: 12/11/2024] Open
Abstract
Pancreatic ductal adenocarcinoma stands out as an exceptionally fatal cancer owing to the complexities associated with its treatment and diagnosis, leading to a notably low five-year survival rate. This study offers a detailed exploration of epidemiological trends in pancreatic cancer and key molecular drivers, such as mutations in CDKN2A, KRAS, SMAD4, and TP53, along with the influence of cancer-associated fibroblasts (CAFs) on disease progression. In particular, we focused on the pivotal roles of signaling pathways such as the transforming growth factor-β and Wnt/β-catenin pathways in the development of pancreatic cancer and investigated their application in emerging therapeutic strategies. This study provides new scientific perspectives on pancreatic cancer treatment, especially in the development of precision medicine and targeted therapeutic strategies, and demonstrates the importance of signaling pathway research in the development of effective therapeutic regimens. Future studies should explore the subtypes of CAFs and their specific roles in the tumor microenvironment to devise more effective therapeutic methods.
Collapse
Affiliation(s)
- Zhong-Yuan Hu
- First School of Clinical Medicine, Shaanxi University of Chinese Medicine, Xianyang 712000, Shaanxi Province, China
| | - Ding Ding
- First School of Clinical Medicine, Shaanxi University of Chinese Medicine, Xianyang 712000, Shaanxi Province, China
| | - Yu Song
- College of Acupuncture and Massage, Shaanxi University of Chinese Medicine, Xianyang 712000, Shaanxi Province, China
| | - Ya-Feng Deng
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou 510000, Guangdong Province, China
| | - Cheng-Ming Zhang
- Digestive Department I, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi’an 710000, Shaanxi Province, China
| | - Tao Yu
- Digestive Department I, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi’an 710000, Shaanxi Province, China
| |
Collapse
|
37
|
Nasiri F, Safarzadeh Kozani P, Salem F, Mahboubi Kancha M, Dashti Shokoohi S, Safarzadeh Kozani P. Mechanisms of antigen-dependent resistance to chimeric antigen receptor (CAR)-T cell therapies. Cancer Cell Int 2025; 25:64. [PMID: 39994651 PMCID: PMC11849274 DOI: 10.1186/s12935-025-03697-y] [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: 10/08/2024] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
Cancer immunotherapy has reshaped the landscape of cancer treatment over the past decades. Genetic manipulation of T cells to express synthetic receptors, known as chimeric antigen receptors (CAR), has led to the creation of tremendous commercial and therapeutic success for the treatment of certain hematologic malignancies. However, since the engagement of CAR-T cells with their respective antigens is solely what triggers their cytotoxic reactions against target cells, the slightest changes to the availability and/or structure of the target antigen often result in the incapacitation of CAR-T cells to enforce tumoricidal responses. This results in the resistance of tumor cells to a particular CAR-T cell therapy that requires meticulous heeding to sustain remissions in cancer patients. In this review, we highlight the antigen-dependent resistance mechanisms by which tumor cells dodge being recognized and targeted by CAR-T cells. Moreover, since substituting the target antigen is the most potent strategy for overcoming antigen-dependent disease relapse, we tend to highlight the current status of some target antigens that might be considered suitable alternatives to the currently available antigens in various cancers. We also propose target antigens whose targeting might reduce the off-tumor adverse events of CAR-T cells in certain malignancies.
Collapse
Affiliation(s)
- Fatemeh Nasiri
- Department of Internal Medicine, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran
| | - Pouya Safarzadeh Kozani
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran.
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Faeze Salem
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maral Mahboubi Kancha
- Faculty of Engineering and Science, School of Science, University of Greenwich, Chatham Maritime, Chatham, Kent, ME4 4TB, UK
| | | | - Pooria Safarzadeh Kozani
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran.
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
38
|
Li J, Liu J, Wu Y, Sun Y, Huang G, Jin M. α-Hederin inhibited pancreatic cancer cell malignant progression by inhibiting LDHA-mediated glycolysis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03621-7. [PMID: 39969605 DOI: 10.1007/s00210-024-03621-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/08/2024] [Indexed: 02/20/2025]
Abstract
α-Hederin is a pentacyclic triterpenoid saponin extracted from Pulsatilla chinensis, which is known to suppress cancer cell proliferation. However, the role of this compound in pancreatic cancer cells remains unclear. The aim of this study was to reveal the docking molecular and the regulatory mechanism of α-hederin in pancreatic cancer. Here, we cultured Capan-1 and BxPC-3 cells and treated with different doses of α-hederin. Cell proliferation, migration, and apoptosis were detected using CCK8, EdU, Transwell, wound healing assay, and flow cytometer apoptosis assay. The in vivo experiment using subcutaneous tumor and caudal vein metastasis model to evaluate the inhibit effect of α-hederin Capan-1 cell tumor growth and metastasis. Proteomics were used to reveal the regulatory mechanism. The result shows that α-hederin treatment inhibits cell proliferation and invasion in concentration dependence way in both vivo and in vitro. The result shows that the IC50 for both Capan-1 and BxPC-3 were 32.5 Mµ and 15 Mµ, respectively. Flow cytometer apoptosis assay shows that α-hederin treatment promotion cell apoptosis in both Capan-1 and BxPC-3 cells. Proteomics and immunofluorescence detection confirmed that α-hederin treatment downregulated lactate dehydrogenase A (LDHA) expression and inhibited glycolysis. Molecular docking of α-hederin and LDHA proteins further confirmed that LDHA is a target of α-hederin. Taken together, this study confirms that α-hederin inhibits pancreatic cancer cell proliferation and invasion by inhibiting LDHA-mediated glycolysis. LDHA may be a direct target of α-hederin in pancreatic cancer.
Collapse
Affiliation(s)
- Jingjing Li
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Jiao Liu
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Yue Wu
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Yi Sun
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, No.1111, XianXia Road, Shanghai, 200336, China.
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| | - Mingming Jin
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| |
Collapse
|
39
|
Maeng JE, Kim JH, Kim SC, Yun WG, Kwon W, Han Y, Oh DY, Lee SH, Jang JY, Ku JL. Comprehensive molecular analysis of 26 newly established human pancreatic ductal adenocarcinoma cell lines reveals two clusters with variating drug sensitivities. Cancer Cell Int 2025; 25:53. [PMID: 39972450 PMCID: PMC11837577 DOI: 10.1186/s12935-025-03671-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 02/01/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a malignant form of cancer with the worst survival rate and an extremely low rate of response to treatments. The development and molecular characterization of pancreatic cancer cell lines (PCCLs) are essential for studying the biology of highly aggressive pancreatic adenocarcinoma. METHODS We applied whole exome sequencing (WES) and RNA-seq to identify molecular characteristics of 26 newly established PCCLs. Eighteen clinically relevant anti-cancer drugs were used to assess highly heterogeneous drug responses across the 26 cell lines. RESULTS We confirmed that common driver mutations of PDAC were well retained in our cell lines through WES analysis. Transcriptomic analysis identified two representative clusters that correlated with responses to certain drugs. By using Moffitt's classification method, the two clusters, C1 and C2, showed comparable expression patterns to "Basal-like" and "Classical" types, respectively. Drug screening results showed varying responses among different cell lines. In our cohort, C2 displayed greater sensitivity to anti-cancer drugs compared to C1. Furthermore, drugs targeting similar molecular pathways exhibited corresponding reactions among cell lines. CONCLUSIONS Our results underscored that transcriptomic features of pancreatic cancer correlate with drug sensitivity rather than with the effects of targeted drugs. Cell lines are useful in vitro model systems for studying the molecular mechanisms of PDAC.
Collapse
Affiliation(s)
- Ju Eun Maeng
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
- Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 101, Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
| | - Jae-Hyeon Kim
- Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 101, Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
| | - Soon-Chan Kim
- Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 101, Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Won-Gun Yun
- Department of Surgery, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
| | - Wooil Kwon
- Department of Surgery, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
| | - Youngmin Han
- Department of Surgery, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Korea
| | - Sang Hyub Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Korea
| | - Jin-Young Jang
- Department of Surgery, Seoul National University College of Medicine, 103 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea.
| | - Ja-Lok Ku
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.
- Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, 101, Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea.
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Korea.
| |
Collapse
|
40
|
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
|
41
|
Wu Z, Zeng L, Fang Z, Yuan Y, Zhou Y, Chen R. Life's Essential 8, genetic susceptibility, and risk of incident pancreatic cancer: A prospective cohort study. Int J Cancer 2025; 156:566-574. [PMID: 39279141 DOI: 10.1002/ijc.35184] [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: 05/21/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/18/2024]
Abstract
The association between the American Heart Association (AHA) Life's Essential 8 (LE8) and the risk of pancreatic cancer (PC) remains unclear. Our goal was to assess the relationships between LE8, genetic susceptibility, and PC risk. This cohort consisted of 234,102 participants from the UK Biobank. The components of LE8 include diet, nicotine exposure, sleep, physical activity, blood glucose, body mass index, blood lipids, and blood pressure. LE8 is classified into three categories: low cardiovascular health (CVH), moderate CVH, and high CVH. Measurements were made using Cox proportional risk models to estimate impact of associations between LE8, genetic susceptibility, and incidence of PC in participants. Compared to participants with low LE8 scores, those with moderate and high LE8 scores had a 53% (HR, 0.47; 95% CI, 0.39-0.57) and 70% (HR, 0.30; 95% CI, 0.22-0.41) lower risk of developing PC, respectively. Interestingly, among individuals with high genetic risk, high LE8 scores were associated with greater benefits (HR, 0.24; 95% CI, 0.15-0.40), whereas the protective effect was weaker among those with low genetic risk (HR, 0.40; 95% CI, 0.21-0.75). Participants with a high LE8 score and a low polygenic risk score (PRS) had the lowest risk of PC (HR, 0.19; 95% CI: 0.11-0.33). Furthermore, we observed a significant additive interaction between LE8 and PRS. A higher LE8 score is associated with a lower risk of PC, especially for participants with a high PRS. These findings have important implications for participants most genetically predisposed to PC and for targeted strategies for PC prevention.
Collapse
Affiliation(s)
- Zhuo Wu
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Liangtang Zeng
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhou Fang
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yuan Yuan
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yu Zhou
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Rufu Chen
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
42
|
Nian Q, Lin Y, Zeng J, Zhang Y, Liu R. Multifaceted functions of the Wilms tumor 1 protein: From its expression in various malignancies to targeted therapy. Transl Oncol 2025; 52:102237. [PMID: 39672002 PMCID: PMC11700300 DOI: 10.1016/j.tranon.2024.102237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/28/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024] Open
Abstract
Wilms tumor 1 (WT1) is a multifaceted protein with dual functions, acting both as a tumor suppressor and as a transcriptional activator of oncogenes. WT1 is highly expressed in various types of solid tumors and leukemia, and its elevated expression is associated with a poor prognosis for patients. High WT1 expression also indicates a greater risk of refractory disease or relapse. Consequently, targeting WT1 is an effective strategy for disease prevention and relapse mitigation. Substantial information is available on the pathogenesis of WT1 in various diseases, and several WT1-targeted therapies, including chemical drugs, natural products, and targeted vaccines, are available. We provide a comprehensive review of the mechanisms by which WT1 influences malignancies and summarize the resulting therapeutic approaches thoroughly. This article provides information on the roles of WT1 in the pathogenesis of different cancers and provides insights into drugs and immunotherapies targeting WT1. The goal of this work is to provide a systematic understanding of the current research landscape and of future directions for WT1-related studies.
Collapse
Affiliation(s)
- Qing Nian
- Department of Transfusion, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32W. Sec. 2, 1st Ring Rd., Qingyang District, Chengdu, Sichuan, China, 610072.
| | - Yan Lin
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, 39 Shierqiaolu, Chengdu, Sichuan, China, 610072
| | - Jinhao Zeng
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, 39 Shierqiaolu, Chengdu, Sichuan, China, 610072
| | - Yanna Zhang
- Department of Transfusion, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32W. Sec. 2, 1st Ring Rd., Qingyang District, Chengdu, Sichuan, China, 610072
| | - Rongxing Liu
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 183 Xinqiao Road, Chongqing, China, 400000.
| |
Collapse
|
43
|
Fu L, Zhou E, Li S, Li Z, Wu L, Zhou X, Tian L, Cui B. Elevated SAMD3 expression in T cells predicts improved survival in pancreatic ductal adenocarcinoma patients. Cancer Immunol Immunother 2025; 74:93. [PMID: 39891760 PMCID: PMC11787060 DOI: 10.1007/s00262-025-03948-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma (PDAC) has an immune-suppressive tumor microenvironment that contributes to resistance to immunotherapy. This study aimed to demonstrate that elevated sterile alpha motif domain-containing protein 3 (SAMD3) expression in effector CD8+ T cells was associated with improved survival in PDAC patients. DESIGN We investigated the heterogeneity and gene expression profiles of T cells using a single-cell RNA sequencing (sc-RNA-seq) dataset comprised of human PDAC samples. SAMD3 mRNA expression was further evaluated in a tumor-specific OT-I/ovalbumin (OVA) mouse model. SAMD3 levels and their clinical significance were evaluated via immunohistochemistry (IHC) analysis. RESULTS SAMD3 was highly expressed in cytotoxic CD8+ T cells, with expression significantly downregulated during T cell exhaustion. SAMD3 levels were positively correlated with CD8+ T cell function. In PDAC patients, high SAMD3 levels in T cells were associated with improved overall survival (OS), disease-free survival (DFS), and T cell infiltration. A patient exhibiting partial response to combination immunotherapy also showed high SAMD3 levels in T cells. CONCLUSION SAMD3 is a biomarker of T cell function, with elevated expression in T cells predicting improved survival. These findings highlight the potential of precision immunotherapy approaches for treating PDAC.
Collapse
Affiliation(s)
- Lingyi Fu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Enliang Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Pancreaticobiliary Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Shuo Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ziteng Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Liyan Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xinxin Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Pancreaticobiliary Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Lang Tian
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Pancreaticobiliary Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Bokang Cui
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Department of Pancreaticobiliary Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
| |
Collapse
|
44
|
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
|
45
|
Mowoe MO, Allam H, Nqada J, Bernon M, Gandhi K, Burmeister S, Kotze U, Kahn M, Kloppers C, Dharshanan S, Azween Z, Maimela P, Townsend P, Jonas E, Blackburn JM. Identification and validation of a novel autoantibody biomarker panel for differential diagnosis of pancreatic ductal adenocarcinoma. Front Immunol 2025; 16:1494446. [PMID: 39949781 PMCID: PMC11821970 DOI: 10.3389/fimmu.2025.1494446] [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: 09/10/2024] [Accepted: 01/07/2025] [Indexed: 02/16/2025] Open
Abstract
Introduction New biomarkers are urgently needed to detect pancreatic ductal adenocarcinoma (PDAC) at an earlier stage for individualized treatment strategies and to improve outcomes. Autoantibodies (AAbs) in principle make attractive biomarkers as they arise early in disease, report on disease-associated perturbations in cellular proteomes, and are static in response to other common stimuli, yet are measurable in the periphery, potentially well in advance of the onset of clinical symptoms. Methods Here, we used high-throughput, custom cancer antigen microarrays to identify a clinically relevant autoantibody biomarker combination able to differentially detect PDAC. Specifically, we quantified the serological AAb profiles of 94 PDAC, chronic pancreatitis (CP), other pancreatic- (PC) and prostate cancers (PRC), non-ulcer dyspepsia patients (DYS), and healthy controls (HC). Results Combinatorial ROC curve analysis on the training cohort data from the cancer antigen microarrays identified the most effective biomarker combination as CEACAM1-DPPA2-DPPA3-MAGEA4-SRC-TPBG-XAGE3 with an AUC = 85·0% (SE = 0·828, SP = 0·684). Additionally, differential expression analysis on the samples run on the iOme™ array identified 4 biomarkers (ALX1-GPA33-LIP1-SUB1) upregulated in PDAC against diseased and healthy controls. Identified AAbs were validated in silico using public immunohistochemistry datasets and experimentally using a custom PDAC protein microarray comprising the 11 optimal AAb biomarker panel. The clinical utility of the biomarker panel was tested in an independent cohort comprising 223 PDAC, PC, PRC, colorectal cancer (CRC), and HC samples. Combinatorial ROC curve analysis on the validation data identified the most effective biomarker combination to be CEACAM1-DPPA2-DPPA3-MAGEA4-SRC-TPBG-XAGE3 with an AUC = 85·0% (SE = 0·828, SP = 0·684). Subsequently, the specificity of the 11-biomarker panel was validated against other cancers (PDAC vs PC: AUC = 70·3%; PDAC vs CRC: AUC = 84·3%; PDAC vs PRC: AUC = 80·2%) and healthy controls (PDAC vs HC: AUC = 80·9%), confirming that this novel AAb biomarker panel is able to selectively detect PDAC amongst other confounding diseases. Conclusion This AAb panel may therefore have the potential to form the basis of a novel diagnostic test for PDAC.
Collapse
Affiliation(s)
- Metoboroghene O Mowoe
- Department of Integrative Biomedical Sciences, Division of Chemical and Systems Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Hisham Allam
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Joshua Nqada
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Marc Bernon
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Karan Gandhi
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Sean Burmeister
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Urda Kotze
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Miriam Kahn
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Christo Kloppers
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Suba Dharshanan
- Recombinant Protein Facility, Sengenics Corporation, Kuala Lumpur, Malaysia
| | - Zafirah Azween
- Recombinant Protein Facility, Sengenics Corporation, Kuala Lumpur, Malaysia
| | - Pamela Maimela
- Department of Integrative Biomedical Sciences, Division of Chemical and Systems Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Paul Townsend
- Faculty of Health Sciences and Sports, University of Stirling, Stirling, United Kingdom
| | - Eduard Jonas
- Surgical Gastroenterology Unit, Division of General Surgery, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Jonathan M Blackburn
- Department of Integrative Biomedical Sciences, Division of Chemical and Systems Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
46
|
Zhang XJ, Lin FF, Wen YQ, Guan KP. Improving molecular subtypes and prognosis of pancreatic cancer through multi group analysis and machine learning. Discov Oncol 2025; 16:96. [PMID: 39873820 PMCID: PMC11775367 DOI: 10.1007/s12672-025-01841-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/20/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PAC) has a complex tumor immune microenvironment, and currently, there is a lack of accurate personalized treatment. Establishing a novel consensus machine learning driven signature (CMLS) that offers a unique predictive model and possible treatment targets for this condition was the goal of this study. METHODS This study integrated multiple omics data of PAC patients, applied ten clustering techniques and ten machine learning approaches to construct molecular subtypes for PAC, and created a new CMLS. RESULTS Using multi-omics clustering, we discovered two cancer subtypes (CSs) associated with prognosis, among which CS1 exhibited poor prognostic outcomes. Subsequently, 13 central genes were identified through screening, constituting CMLS with a significant prognostic ability. The low CMLS group had a better prognosis and was more likely to possess a "hot" tumor phenotype. The prognosis for the high CMLS group was dismal. Still, the tumor mutation burden (TMB) and tumor neoantigen burden (TNB) levels in this group of patients were higher than in the low CMLS group, which were more favorable for immune therapy response. CONCLUSION This study emphasizes that CMLS provides a beneficial instrument for early prediction of patient prognosis and screening of probable patients appropriate for immunotherapy and has broad implications for clinical practice.
Collapse
Affiliation(s)
- Xue-Jian Zhang
- Department of Laboratory, the Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Fang-Fang Lin
- Department of Laboratory, the Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Ya-Qing Wen
- Department of Laboratory, the Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Kun-Ping Guan
- Department of Laboratory, the Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China.
| |
Collapse
|
47
|
Bai Z, Xia Q, Xu W, Wu Z, He X, Zhang X, Wang Z, Luo M, Sun H, Liu S, Wang J. N 6-Methylandenosine-related lncRNAs as potential biomarkers for predicting prognosis and the immunotherapy response in pancreatic cancer. Cell Mol Life Sci 2025; 82:48. [PMID: 39833465 PMCID: PMC11753445 DOI: 10.1007/s00018-024-05573-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 01/22/2025]
Abstract
Emerging evidence has shown that the N6-methyladenosine (m6A) modification of RNA plays key roles in tumorigenesis and the progression of various cancers. However, the potential roles of the m6A modification of long noncoding RNAs (lncRNAs) in pancreatic cancer (PaCa) are still unknown. To analyze the prognostic value of m6A-related lncRNAs in PaCa, an m6A-related lncRNA signature was constructed as a risk model via Pearson's correlation and univariate Cox regression analyses in The Cancer Genome Atlas (TCGA) database. The tumor microenvironment (TME), tumor mutation burden, and drug sensitivity of PaCa were investigated by m6A-related lncRNA risk score analyses. We established an m6A-related risk prognostic model consisting of five lncRNAs, namely, LINC01091, AC096733.2, AC092171.5, AC015660.1, and AC005332.6, which not only revealed significant differences in immune cell infiltration associated with the TME between the high-risk and low-risk groups but also predicted the potential benefit of immunotherapy for patients with PaCa. Drugs such as WZ8040, selumetinib, and bortezomib were also identified as more effective for high-risk patients. Our results indicate that the m6A-related lncRNA risk model could be an independent prognostic indicator, which may provide valuable insights for identifying therapeutic approaches for PaCa.
Collapse
Affiliation(s)
- Zhihui Bai
- Central Laboratory, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, China
- Xiamen Key Laboratory of Biotherapy, Xiamen, 361015, China
| | - Qianlin Xia
- Laboratory Medicine, Shanghai Sixth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Wanli Xu
- Central Laboratory, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, China
| | - Zhirong Wu
- Department of General Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, China
| | - Xiaomeng He
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, China
| | - Xin Zhang
- Central Laboratory, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, China
| | - Zhefeng Wang
- Xiamen Key Laboratory of Biotherapy, Xiamen, 361015, China
- Clinical Research Center for Precision Medicine of Abdominal Tumor of Fujian Province, Xiamen, China
| | - Mengting Luo
- Central Laboratory, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, China
| | - Huaqin Sun
- Central Laboratory, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, China
| | - Songmei Liu
- Department of Clinical Laboratory, Center for Gene Diagnosis & Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jin Wang
- Central Laboratory, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, China.
- Xiamen Key Laboratory of Biotherapy, Xiamen, 361015, China.
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, China.
| |
Collapse
|
48
|
Yousef M, Hurd MW, Yousef A, Ludmir EB, Pillai AB, Peterson J, Koay EJ, Albarouki S, Tzeng CW, Snyder R, Katz MHG, Wang H, Overman MJ, Maitra A, Pant S, Smaglo BG, Wolff RA, Yao J, Shen JP, Zhao D. Clinical and molecular characteristics of patients with brain metastasis secondary to pancreatic ductal adenocarcinoma. Oncologist 2025; 30:oyae182. [PMID: 39014543 PMCID: PMC11783327 DOI: 10.1093/oncolo/oyae182] [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: 04/23/2024] [Accepted: 06/21/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND The prognosis for patients with pancreatic ductal adenocarcinoma (PDAC) is poor. Secondary brain metastasis (Br-M) occurs in less than 1% of patients. Clinical characteristics and molecular alterations have not been characterized in this rare patients' subset. MATERIALS AND METHODS The Foundry software platform was used to retrospectively query electronic health records for patients with Br-M secondary to PDAC from 2005 to 2023; clinical, molecular, and overall survival (OS) data were analyzed. RESULTS Br-M was diagnosed in 44 patients with PDAC. Median follow-up was 78 months; median OS from initial PDAC diagnosis was 47 months. Median duration from PDAC diagnosis to Br-M detection was 24 months; median OS from Br-M diagnosis was 3 months. At Br-M diagnosis, 82% (n = 36) of patients had elevated CA19-9. Lung was the most common preexisting metastatic location (71%) with Br-M, followed by liver (66%). Br-M were most frequently observed in the frontal lobe (34%, n = 15), cerebellar region (23%, n = 10), and leptomeninges (18%, n = 8). KRAS mutations were detected in 94.1% (n = 16) of patients who had molecular data available (n = 17) with KRASG12V being the most frequent subtype 47% (n = 8); KRASG12D in 29% (n = 5); KRASG12R in 18% (n = 3). Patients who underwent Br-M surgical resection (n = 5) had median OS of 8.6 months, while median OS following stereotactic radiosurgery only (n = 11) or whole-brain radiation only (n = 20) was 3.3 and 2.8 months, respectively. CONCLUSION Br-M is a late PDAC complication, resulting in an extremely poor prognosis especially in leptomeningeal disease. KRAS was mutated in 94.1% of the patients and the KRASG12V subtype was prevalent.
Collapse
Affiliation(s)
- Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mark W Hurd
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Abdelrahman Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ethan B Ludmir
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ashwathy B Pillai
- Department of Hospital Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jennifer Peterson
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Eugene J Koay
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sali Albarouki
- Department of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, United States
| | - Ching-Wei Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rebecca Snyder
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Huamin Wang
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anirban Maitra
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Brandon G Smaglo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James Yao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - John P Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dan Zhao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
49
|
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: 2] [Impact Index Per Article: 2.0] [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
|
50
|
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
|