1
|
Huang Y, Chen L, Chen Y, Zhou S, Xie X, Xie J, Yu M, Chen J. High-density lipoprotein-based nanoplatform reprograms tumor microenvironment and enhances chemotherapy against pancreatic ductal adenocarcinoma. Biomaterials 2025; 318:123147. [PMID: 39908877 DOI: 10.1016/j.biomaterials.2025.123147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/25/2024] [Accepted: 01/26/2025] [Indexed: 02/07/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly aggressive, with limited success in traditional therapies due to the fibrotic, immunosuppressive, pro-metastatic tumor microenvironment (TME), which collectively impede the drug accumulation and accelerate the tumor progression. In this work, we developed a PDAC-customized nutrient-mimicking reconstituted high-density lipoprotein (rHDL) capable of efficiently co-encapsulate versatile TME regulating cannabidiol and cytotoxic gemcitabine to simultaneously reprogram TME while suppressing PDAC progression. Specifically, a small-sized, nutrient-like rHDL was constructed to realize deep PDAC parenchyma penetration and efficient intra-tumoral uptake. Next, natural herbal compound cannabidiol was screened and incorporated into rHDL to regulate TME via attenuating fibrosis, reliving immunosuppression and mitigating metastatic tendency. At last, gemcitabine, the PDAC gold standard first-line therapy was co-delivered by the PDAC-customized rHDL to overcome drug resistance and amplify its PDAC suppression. Our findings demonstrate the feasibility of an integrated multi-stage TME regulation strategy for improved PDAC therapy, and might represent a modality in promoting chemotherapy against PDAC.
Collapse
Affiliation(s)
- Yukun Huang
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China; Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liang Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Yu Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Songlei Zhou
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Xiaoying Xie
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Jing Xie
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Minghua Yu
- Fudan University Clinical Research Center for Cell-based Immunotherapy & Department of Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Shanghai, 201399, China
| | - Jun Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
2
|
Vats M, Rathod D, Patel H, Richards T, Patel K. Self-emulsifying Nano-PND oral delivery systems of PND1186: In silico modeling for bioavailability estimation. J Mol Liq 2025; 426:127161. [PMID: 40322757 PMCID: PMC12048016 DOI: 10.1016/j.molliq.2025.127161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Focal adhesion kinase (FAK) inhibitors have proven to aid the therapeutic potential of anti-cancer agents. PND1186 (PND) is a FAK inhibitor disrupting the oncogenic processes such as cell survival, proliferation, adhesion, migration and angiogenesis, as well as remodeling of tumor microenvironment. However, the pharmacological potential of PND is limited by its poor solubility and bioavailability due to rapid precipitation of weakly basic PND in the intestinal milieu. As a solution, we have developed a self-nanoemulsifying PND oral delivery system (NanoPODS) for rapid dissolution of PND while Soluplus containing system (NanoPODS-S) was prepared to prevent the precipitation of PND. Optimized NanoPODS-S depicted a particle size of 107.0 ± 3.6 nm, PDI of 0.223 ± 0.016, and a surface potential of -4.2 ± 0.007 mV, along with > 70% PND released at pH 6.8. In silico pharmacokinetics predicted 99% oral bioavailability for NanoPODS-S. This study evaluates the efficacy of NanoPODS and NanoPODS-S for improved oral bioavailability with better cytotoxicity efficacy on Pancreatic Ductal Adenocarcinoma (PDAC) cell lines. NanoPODS-S is the first of its kind, self-nanoemulsifying system containing a polymeric precipitation inhibitor mimicking a "spring-parachute effect". It will be a novel platform technology for rapid and enhanced dissolution of poorly soluble molecules.
Collapse
Affiliation(s)
- Mukti Vats
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| | - Drishti Rathod
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| | - Henis Patel
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| | - Terjahna Richards
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| | - Ketan Patel
- College of Pharmacy and Health Sciences, St. John’s University, NY, USA
| |
Collapse
|
3
|
Zhao K, Muralidharan V, Brown S, Upton A, Alshimirti M, Cooray PD. Neoadjuvant Pembrolizumab Enables Successful Downstaging and Resection of Borderline Resectable MSI-H/dMMR Pancreatic Ductal Adenocarcinoma: A Case Report and Literature Review. J Gastrointest Cancer 2025; 56:112. [PMID: 40341577 PMCID: PMC12062155 DOI: 10.1007/s12029-025-01237-5] [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] [Accepted: 04/29/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with a poor prognosis. While immunotherapy has shown limited efficacy in most PDAC cases due to an immunosuppressive tumour microenvironment, tumours with microsatellite instability-high (MSI-H) or deficient mismatch repair (dMMR) status exhibit increased responsiveness to immune checkpoint inhibitors. CASE PRESENTATION We report the case of a 45-year-old woman with Lynch syndrome who was diagnosed with MSI-H/dMMR PDAC during routine surveillance. Given the borderline resectable nature of her tumour and previous chemotherapy-related neurotoxicity, she was treated with neoadjuvant pembrolizumab instead of conventional chemotherapy. Following four cycles of pembrolizumab, imaging revealed a marked metabolic response, allowing for successful R0 pancreatoduodenectomy. Postoperative histology confirmed a significant reduction in tumour size, and immunohistochemical analysis demonstrated increased CD8 + T cell infiltration, supporting an enhanced anti-tumour immune response. The patient continues adjuvant pembrolizumab therapy without complications. CONCLUSION This case highlights the potential role of neoadjuvant pembrolizumab in MSI-H/dMMR PDAC, demonstrating successful tumour downstaging and facilitating surgical resection. Our findings support further investigation into the integration of immunotherapy as a neoadjuvant strategy for select PDAC patients.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Female
- Middle Aged
- Neoadjuvant Therapy/methods
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/surgery
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/surgery
- Pancreatic Neoplasms/genetics
- Microsatellite Instability
- DNA Mismatch Repair
- Antineoplastic Agents, Immunological/therapeutic use
- Pancreaticoduodenectomy
- Colorectal Neoplasms, Hereditary Nonpolyposis
Collapse
Affiliation(s)
- Kevin Zhao
- University of Melbourne School of Medicine, Melbourne, Australia
| | - Vijayaragavan Muralidharan
- Prometheus Research Collaborative, Department of Surgery, Austin Precinct, The University of Melbourne, Austin Health, Melbourne, Australia
| | | | | | | | - Prasad D Cooray
- Department of Surgery, The University of Melbourne, Austin Health, Melbourne, Australia.
| |
Collapse
|
4
|
Byun JH, Lebeau PF, Trink J, Uppal N, Lanktree MB, Krepinsky JC, Austin RC. Endoplasmic reticulum stress as a driver and therapeutic target for kidney disease. Nat Rev Nephrol 2025; 21:299-313. [PMID: 39988577 DOI: 10.1038/s41581-025-00938-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 02/25/2025]
Abstract
The endoplasmic reticulum (ER) has crucial roles in metabolically active cells, including protein translation, protein folding and quality control, lipid biosynthesis, and calcium homeostasis. Adverse metabolic conditions or pathogenic genetic variants that cause misfolding and accumulation of proteins within the ER of kidney cells initiate an injurious process known as ER stress that contributes to kidney disease and its cardiovascular complications. Initiation of ER stress activates the unfolded protein response (UPR), a cellular defence mechanism that functions to restore ER homeostasis. However, severe or chronic ER stress rewires the UPR to activate deleterious pathways that exacerbate inflammation, apoptosis and fibrosis, resulting in kidney injury. This insidious crosstalk between ER stress, UPR activation, oxidative stress and inflammation forms a vicious cycle that drives kidney disease and vascular damage. Furthermore, genetic variants that disrupt protein-folding mechanisms trigger ER stress, as evidenced in autosomal-dominant tubulointerstitial kidney disease and Fabry disease. Emerging therapeutic strategies that enhance protein-folding capacity and reduce the burden of ER stress have shown promising results in kidney diseases. Thus, integrating knowledge of how genetic variants cause protein misfolding and ER stress into clinical practice will enhance treatment strategies and potentially improve outcomes for various kidney diseases and their vascular complications.
Collapse
Affiliation(s)
- Jae Hyun Byun
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Paul F Lebeau
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Jackie Trink
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Nikhil Uppal
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine and Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Matthew B Lanktree
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine and Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Joan C Krepinsky
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Richard C Austin
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada.
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
5
|
Pele KG, Calderón-Villalba A, Amaveda H, Mora M, Zhang-Zhou J, Pérez MÁ, García-Aznar JM, Alamán-Díez P, García-Gareta E. Novel hydrogel-based cancer-on-a-chip models for growth of 3D multi-cellular structures and investigation of early angiogenesis in pancreatic ductal adenocarcinoma. Colloids Surf B Biointerfaces 2025; 253:114736. [PMID: 40315572 DOI: 10.1016/j.colsurfb.2025.114736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/27/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
Cancer-on-a-chip models have enormous potential for the study of tumour development events. Here, we investigated hydrogels of egg white (EW) and gelatin for growth of 3D multi-cellular structures and investigation of early angiogenesis inside microfluidic devices. We focused on pancreatic ductal adenocarcinoma (PDAC), a devastating gastrointestinal malignancy. EW/gelatin hydrogels were stiffer and showed porous globular structures compared to the fibrous network of collagen I molecules. PANC-1 cells preferentially formed significantly larger spheroids in collagen I than in EW/gelatin hydrogels, whilst cell aggregates in the shape of grape-like clusters were significantly larger and more abundant in EW/gelatin. Cells inside the aggregates showed active cell unions, secreted matrix, and formed active unions with the surrounding EW/gelatin hydrogel. Early stages of PDAC were recreated by co-culture of two different microenvironments, one for PANC-1 and another one for fibroblasts, for investigating the secretion of soluble angiogenic factors, which depended on the role of each factor in the angiogenic and tumorigenic processes. Overall, cancer cell proliferation and establishment of a tumour vasculature were favoured. This study demonstrates the importance of the microenvironment in tumour cells behaviour as well as the complex interplay between the different cells present in PDAC to establish a tumoural vasculature.
Collapse
Affiliation(s)
- Karinna Georgiana Pele
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Alejandro Calderón-Villalba
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Hippolyte Amaveda
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC and University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Mario Mora
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC and University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Jack Zhang-Zhou
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - María Ángeles Pérez
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain
| | - José Manuel García-Aznar
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain
| | - Pilar Alamán-Díez
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Elena García-Gareta
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain; Division of Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, University College London, London NW3 2QG, United Kingdom.
| |
Collapse
|
6
|
Liu W, Fang Y, Zhang C, Xiang M, Qi L, Su A, Sun Y. Cost-effectiveness of NALIRIFOX versus Nab-paclitaxel and gemcitabine in previously untreated metastatic pancreatic ductal adenocarcinoma. BMC Gastroenterol 2025; 25:266. [PMID: 40247200 PMCID: PMC12004809 DOI: 10.1186/s12876-025-03867-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND & AIMS The NAPOLI 3 trial demonstrated that compared to the nab-paclitaxel and gemcitabine (GemNab) regimen, the NALIRIFOX (5-fluorouracil, leucovorin, liposomal irinotecan, oxaliplatin) regimen can significantly improve patients' overall survival (OS) and progression-free survival (PFS), and the safety of this regimen is generally controllable. To ensure the appropriateness of the chosen treatment, economic evaluation is essential. Thus, from the perspective of American healthcare systems, we explored the cost-effectiveness comparison between NALIRIFOX or GemNab in treating patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) who had not previously received treatment. METHODS A three-state partitioned survival model was developed, incorporating a lifetime horizon with a 4-week cycle length. The efficacy data were sourced from the NAPOLI 3 study, while treatment costs, health state utilities, and adverse events (AEs) were obtained from public databases and literature. Applying an annual discount rate of 3%, the analysis focused on total cost, quality-adjusted life-years (QALYs), and the incremental cost-effectiveness ratio (ICER) as primary outcomes. Conducted within a US healthcare system perspective over a 13-year lifetime horizon, the analysis set willingness-to-pay (WTP) thresholds of $50,000, $100,000 and $150,000 per QALY. Sensitivity analyses were performed to assess the robustness of the model's findings. RESULTS In our base-case analysis, NALIRIFOX group was estimated to achieve an incremental gain of 0.13 QALYs. Compared to the GemNab group, treatment with NALIRIFOX resulted in higher costs ($150,437 vs $130,683). The ICER for NALIRIFOX therapy was calculated to be $155,602.83/QALYs. Sensitivity analysis revealed the model's robustness across a range of parameters. Through probabilistic sensitivity analysis applying Monte Carlo simulation with 1000 iterations, NALIRIFOX therapy was identified as the cost-effective regimen in 45.7% of all iterations at a WTP threshold of $150,000/QALYs. CONCLUSIONS Treatment with NALIRIFOX strategy was estimated to provide a significant clinical advantage over GemNab. According to the model outcomes, NALIRIFOX therapy is borderline cost-effective for treatment-naive patients with mPDAC, slightly exceeding the highest conventional US willingness-to-pay threshold of 150,000/QALY. However, at lower thresholds, such as 100,000/QALY, NALIRIFOX fails to demonstrate cost-effectiveness in all simulations. In addition, NALIRIFOX therapy emerges as a cost-effective treatment strategy at higher willingness-to-pay thresholds or when the drug price is significantly reduced.
Collapse
Affiliation(s)
- Wenjie Liu
- National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District Panjiayuan Nanli No. 17, Beijing, 100021, China
| | - Yuting Fang
- National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District Panjiayuan Nanli No. 17, Beijing, 100021, China
| | - Chao Zhang
- Department of Medical Oncology, Beijing Chaoyang District Sanhuan Cancer Hospital, Beijing, 100122, China
| | - Midan Xiang
- National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District Panjiayuan Nanli No. 17, Beijing, 100021, China
| | - Lijuan Qi
- National Cancer Center, National Clinical Research Center for Cancer/Hebei Cancer Hospital, Chinese Academy of Medical Sciences, Langfang, 065001, China
| | - Aijiang Su
- Department of Medical Oncology, Beijing Chaoyang District Sanhuan Cancer Hospital, Beijing, 100122, China
| | - Yongkun Sun
- National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District Panjiayuan Nanli No. 17, Beijing, 100021, China.
| |
Collapse
|
7
|
Huang C, Wang T, Chen R, Xu Y. Discovery of CMNPD31124 as a novel marine-derived PKMYT1 inhibitor for pancreatic ductal adenocarcinoma therapy: computational and biological insights. Front Pharmacol 2025; 16:1569765. [PMID: 40290442 PMCID: PMC12022493 DOI: 10.3389/fphar.2025.1569765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/10/2025] [Indexed: 04/30/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest cancers due to its late diagnosis, resistance to therapy, and a dismal 5-year survival rate of only 12%. Overexpression of PKMYT1-a key regulator of the cell cycle-correlates with poor patient outcomes, making it a promising therapeutic target. In this study, we identify CMNPD31124, a novel marine-derived indole alkaloid, as a potent PKMYT1 inhibitor. Molecular docking revealed that CMNPD31124 has superior binding affinity compared to the reference compound Cpd 4, forming robust interactions with critical residues such as CYS-190, TYR-121, and GLY-122. Molecular dynamics simulations further demonstrated its stable binding conformation and dynamic adaptability, with Chai-1 modeling supporting a covalent binding mechanism at the PKMYT1 active site. Importantly, in vitro assays showed that CMNPD31124 exhibits an IC50 of 18.6 μM in MiaPaCa-2 cells and 31.7 μM in BXPC3 cells, while concentrations up to 80 μM did not significantly affect normal pancreatic cells. Despite these promising results, toxicity predictions indicate potential hepatotoxicity and neurotoxicity, highlighting the need for further structural optimization. This work lays a solid foundation for the rational design of PKMYT1 inhibitors by integrating computational methods with insights from marine natural products.
Collapse
Affiliation(s)
- Chaojie Huang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicinesla, Hangzhou, China
| | - Ting Wang
- The Third Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rui Chen
- College of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Yunyun Xu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
8
|
Ulus G, Özbek EN, Yılmaz H, Keselik E, Sarıcaoğlu M, Akyol Bahçeci S, İşel E, Debeleç Bütüner B, Yetik Anacak G, Koparal AT. Borax pentahydrate as a promising boron-based angiogenesis inhibitor. J Trace Elem Med Biol 2025; 89:127640. [PMID: 40203787 DOI: 10.1016/j.jtemb.2025.127640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Boron, a trace element, is involved in various physiological and metabolic processes, and recent studies suggest that boron compounds may have potential in cancer prevention and treatment. In this study, the antiangiogenic effects of a boron compound, borax pentahydrate (BPH), were investigated. Angiogenesis is a tightly regulated biological process responsible for the formation of new blood vessels from existing vasculatures. This process plays a critical role in cancer progression, making it an important target for cancer therapy. Pancreatic and kidney cancers are difficult to treat because they are aggressive and resistant to chemotherapy. METHODS The antiproliferative activity was evaluated using the MTT assay, while antiangiogenic effects were tested through in vitro tube formation assays and in ovo chick chorioallantoic membrane (CAM) assay. The effect of BPH on VEGF levels was determined using Western blot analysis in HUVEC, ACHN, PANC-1 cells. The effect of BPH on tumor angiogenesis was investigated with an in vivo Ehrlich ascites carcinoma model (EAC). RESULTS BPH exhibited potent antiproliferative and antiangiogenic activities, inhibiting the proliferation of ACHN, PANC-1, and HUVECs, disrupting endothelial tube formation, and inhibiting vascular formation on the CAM surface in a dose-dependent manner. VEGF levels were significantly decreased in ACHN, PANC-1 and HUVECs. There was also a decrease in VEGF and TGF-β1 levels in BPH-treated tumor groups. In addition, BPH caused a decrease in tumor size. CONCLUSION These findings suggest that BPH may be a new antiangiogenic and antitumoral agent. BPH may contribute to drug development studies targeting angiogenesis-related diseases as a promising new therapeutic agent.
Collapse
Affiliation(s)
- G Ulus
- Republic of Türkiye, Ministry of Education, Şerife Bacı Vocational and Technical High School, Izmir 35090, Turkiye.
| | - E N Özbek
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkiye
| | - H Yılmaz
- Republic of Türkiye, Ministry of Education, Mimar Sinan Vocational and Technical High School, Izmir 35090, Turkiye
| | - E Keselik
- Department of Histology and Embryology, Faculty of Medicine, Katip Çelebi University, Izmir 35100, Turkiye
| | - M Sarıcaoğlu
- Department of Histology and Embryology, Faculty of Medicine, Katip Çelebi University, Izmir 35100, Turkiye
| | - S Akyol Bahçeci
- Department of Histology and Embryology, Faculty of Medicine, Katip Çelebi University, Izmir 35100, Turkiye
| | - E İşel
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkiye
| | - B Debeleç Bütüner
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkiye
| | - G Yetik Anacak
- Department of Pharmacology, Faculty of Pharmacy, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkiye
| | - A T Koparal
- Yunus Emre Vocational School of Health Services, Anadolu University, Eskisehir 26470, Turkiye
| |
Collapse
|
9
|
Aylar D, Karatug Kacar A. Studies on Treatment Within the Scope of Medical Biotechnology for Pancreatic Diseases. Mol Biotechnol 2025; 67:1321-1335. [PMID: 38627328 DOI: 10.1007/s12033-024-01142-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/14/2024] [Indexed: 03/22/2025]
Abstract
The pancreas is made of two compartments: the exocrine pancreas, a source of digestive enzymes, and the endocrine islets which produce vital hormones. Distinct diseases could arise in the pancreas such as diabetes, neuroendocrine tumors, pancreatitis, and pancreatic cancers. Various treatment methods are being researched against these diseases. Treatment with recombinant proteins, therapeutic antibodies, vaccination, gene therapy, tissue engineering, and stem cell treatment are treatment methods. Furthermore, biomarkers are important for both treatment and diagnosis. However, some of the treatment methods mentioned above have not yet been applied to some pancreatic diseases. This review provides insights into the latest advancements in diagnosis and treatment for pancreatic diseases within the scope of medical biotechnology. In addition, some methods that are not yet used for treatment purposes for pancreatic diseases but are used in other diseases that occur in different organs due to similar reasons have been investigated. In this context, possible diagnosis and treatment methods for pancreatic diseases are interpreted. The first aim of this review is to bring together and present the current diagnosis and treatment methods for pancreatic diseases. The second aim is to highlight methods that may have treatment potential by comparing pancreatic diseases that cannot be treated with similar diseases.
Collapse
Affiliation(s)
- Dilara Aylar
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Ayse Karatug Kacar
- Department of Biology, Faculty of Science, Istanbul University, Vezneciler, Istanbul, Turkey.
| |
Collapse
|
10
|
Zaidh SM, Vengateswaran HT, Habeeb M, Aher KB, Bhavar GB, Irfan N, Lakshmi KNVC. Network pharmacology and AI in cancer research uncovering biomarkers and therapeutic targets for RALGDS mutations. Sci Rep 2025; 15:10938. [PMID: 40157967 PMCID: PMC11954960 DOI: 10.1038/s41598-025-91568-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/21/2025] [Indexed: 04/01/2025] Open
Abstract
The lack of target therapies is accountable for the higher mortality of various types of cancer. To address this issue, we selected a target mutated Kirsten rat sarcoma virus oncogene homologue, which plays a significant role in various cancers. Our study aims to identify selective biomarkers and develop diagnostic and therapeutic strategies for KRAS-associated genes using artificial intelligence. Initially, Genomic data, cancer epidemiology, proteomics network interactions, and omics enrichment were analyzed. Structured E-pharmacophore model aided in capturing the binding cavity using eraser algorithms and fabricating a new selective lead compound for the KRSA. The selective molecule was abridged inside the binding cavity and stability was validated through 100 ns molecular dynamics simulations. Epidemiological-neural network studies indicated KRAS mutations leads 40 types of cancer, exclusively pancreatic and colorectal cancers, with diploid and missense mutations as primary factors. Pathway analysis highlighted the involvement of the MAPK and RAS signaling pathways in cancer development and proteomics analysis identified RALGDS as a key protein. Protein-based pharmacophore analysis mapped the biologically active features such as donor, acceptor and aromatic ring with the designed ligands. The results of interaction interpretation illustrate that the amino acid Tyr566 formed an H-bond interaction with the amine group of the octyl ring system and 20 amino acids crafted to properly orient the molecule to fit inside the polar cavity of KRAS protein. The MMGBSA score of - 53.33 kcal/mol conformed to the well-configured binding with KRSA and realistic model simulation exposed the π-π, π-cationic and hydrophobic interactions stabilised the molecule inside the KRSA protein throughout 100 ns simulation. The study demonstrates the vitality of AI and network pharmacology to identify potential-target biomarkers for KRAS-associated genes, paving the way for improved cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- S Mohammed Zaidh
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India
- D3 Drug Tech Lab Pvt Ltd, Chennai, 600048, India
| | | | - Mohammad Habeeb
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India
| | - Kiran Balasaheb Aher
- Department of Pharmaceutical Quality Assurance, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra, 424001, India
| | - Girija Balasaheb Bhavar
- Department of Pharmaceutical Chemistry, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra, 424001, India
| | - N Irfan
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India.
| | - K N V Chenchu Lakshmi
- Department of Pharmaceutical Chemistry, KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Green Fields, Vaddeswaram, A.P, 522302, India
| |
Collapse
|
11
|
Araújo CC, Frias J, Mendes F, Martins M, Mota J, Almeida MJ, Ribeiro T, Macedo G, Mascarenhas M. Unlocking the Potential of AI in EUS and ERCP: A Narrative Review for Pancreaticobiliary Disease. Cancers (Basel) 2025; 17:1132. [PMID: 40227709 PMCID: PMC11988021 DOI: 10.3390/cancers17071132] [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: 01/24/2025] [Revised: 02/14/2025] [Accepted: 03/03/2025] [Indexed: 04/15/2025] Open
Abstract
Artificial Intelligence (AI) is transforming pancreaticobiliary endoscopy by enhancing diagnostic accuracy, procedural efficiency, and clinical outcomes. This narrative review explores AI's applications in endoscopic ultrasound (EUS) and endoscopic retrograde cholangiopancreatography (ERCP), emphasizing its potential to address diagnostic and therapeutic challenges in pancreaticobiliary diseases. In EUS, AI improves pancreatic mass differentiation, malignancy prediction, and landmark recognition, demonstrating high diagnostic accuracy and outperforming traditional guidelines. In ERCP, AI facilitates precise biliary stricture identification, optimizes procedural techniques, and supports decision-making through real-time data integration, improving ampulla recognition and predicting cannulation difficulty. Additionally, predictive analytics help mitigate complications like post-ERCP pancreatitis. The future of AI in pancreaticobiliary endoscopy lies in multimodal data fusion, integrating imaging, genomic, and molecular data to enable personalized medicine. However, challenges such as data quality, external validation, clinician training, and ethical concerns-like data privacy and algorithmic bias-must be addressed to ensure safe implementation. By overcoming these challenges, AI has the potential to redefine pancreaticobiliary healthcare, improving diagnostic accuracy, therapeutic outcomes, and personalized care.
Collapse
Affiliation(s)
- Catarina Cardoso Araújo
- Precision Medicine Unit, Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal; (C.C.A.); (J.F.); (F.M.); (M.M.); (J.M.); (M.J.A.); (T.R.); (G.M.)
- WGO Gastroenterology and Hepatology Training Center, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
| | - Joana Frias
- Precision Medicine Unit, Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal; (C.C.A.); (J.F.); (F.M.); (M.M.); (J.M.); (M.J.A.); (T.R.); (G.M.)
- WGO Gastroenterology and Hepatology Training Center, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
| | - Francisco Mendes
- Precision Medicine Unit, Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal; (C.C.A.); (J.F.); (F.M.); (M.M.); (J.M.); (M.J.A.); (T.R.); (G.M.)
- WGO Gastroenterology and Hepatology Training Center, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
| | - Miguel Martins
- Precision Medicine Unit, Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal; (C.C.A.); (J.F.); (F.M.); (M.M.); (J.M.); (M.J.A.); (T.R.); (G.M.)
- WGO Gastroenterology and Hepatology Training Center, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
| | - Joana Mota
- Precision Medicine Unit, Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal; (C.C.A.); (J.F.); (F.M.); (M.M.); (J.M.); (M.J.A.); (T.R.); (G.M.)
- WGO Gastroenterology and Hepatology Training Center, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
| | - Maria João Almeida
- Precision Medicine Unit, Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal; (C.C.A.); (J.F.); (F.M.); (M.M.); (J.M.); (M.J.A.); (T.R.); (G.M.)
- WGO Gastroenterology and Hepatology Training Center, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
| | - Tiago Ribeiro
- Precision Medicine Unit, Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal; (C.C.A.); (J.F.); (F.M.); (M.M.); (J.M.); (M.J.A.); (T.R.); (G.M.)
- WGO Gastroenterology and Hepatology Training Center, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
| | - Guilherme Macedo
- Precision Medicine Unit, Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal; (C.C.A.); (J.F.); (F.M.); (M.M.); (J.M.); (M.J.A.); (T.R.); (G.M.)
- WGO Gastroenterology and Hepatology Training Center, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
- Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
| | - Miguel Mascarenhas
- Precision Medicine Unit, Department of Gastroenterology, São João University Hospital, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal; (C.C.A.); (J.F.); (F.M.); (M.M.); (J.M.); (M.J.A.); (T.R.); (G.M.)
- WGO Gastroenterology and Hepatology Training Center, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
- Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-427 Porto, Portugal
| |
Collapse
|
12
|
Jørgensen LV, Christensen EB, Barnkob MB, Barington T. The clinical landscape of CAR NK cells. Exp Hematol Oncol 2025; 14:46. [PMID: 40149002 PMCID: PMC11951618 DOI: 10.1186/s40164-025-00633-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Chimeric antigen receptor (CAR) NK cell therapy has emerged as a promising alternative to CAR T cell therapy, offering significant advantages in terms of safety and versatility. Here we explore the current clinical landscape of CAR NK cells, and their application in hematologic malignancies and solid cancers, as well as their potential for treating autoimmune disorders. Our analysis draws from data collected from 120 clinical trials focused on CAR NK cells, and presents insights into the demographics and characteristics of these studies. We further outline the specific targets and diseases under investigation, along with the major cell sources, genetic modifications, combination strategies, preconditioning- and dosing regimens, and manufacturing strategies being utilized. Initial results from 16 of these clinical trials demonstrate promising efficacy of CAR NK cells, particularly in B cell malignancies, where response rates are comparable to those seen with CAR T cells but with lower rates of severe adverse effects, such as cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and graft-versus-host disease (GvHD). However, challenges remain in solid tumor applications, where only modest efficacy has been observed to date. Our analysis reveals that research is increasingly focused on enhancing CAR NK cell persistence, broadening their therapeutic targets, and refining manufacturing processes to improve accessibility and scalability. With recent advancements in NK cell engineering and their increased clinical applications, CAR NK cells are predicted to become an integral component of next-generation immunotherapies, not only for cancer but potentially for immune-mediated diseases as well.
Collapse
Affiliation(s)
- Lasse Vedel Jørgensen
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense, Denmark
| | - Emil Birch Christensen
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense, Denmark
| | - Mike Bogetofte Barnkob
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense, Denmark
| | - Torben Barington
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark.
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense, Denmark.
| |
Collapse
|
13
|
Moore LL, Houchen CW. Evolving insights into pancreatic tumor initiation and progression through DCLK1-expressing tuft cells. Dev Cell 2025; 60:817-818. [PMID: 40132546 DOI: 10.1016/j.devcel.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
In this issue of Developmental Cell, Salas-Escabillas et al. find that tuft cells undergo a tuft-to-neuroendocrine transition, switching from tumor suppressors to tumor promoters via c-Myc in pancreatic ductal adenocarcinoma (PDAC). This result offers exciting insights into PDAC's aggressiveness, therapy resistance, and high mortality.
Collapse
Affiliation(s)
- Landon L Moore
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | - Courtney W Houchen
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA; The Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
14
|
Guo Y, Morshedi M. Cutting-edge nanotechnology: unveiling the role of zinc oxide nanoparticles in combating deadly gastrointestinal tumors. Front Bioeng Biotechnol 2025; 13:1547757. [PMID: 40182988 PMCID: PMC11966175 DOI: 10.3389/fbioe.2025.1547757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/21/2025] [Indexed: 04/05/2025] Open
Abstract
Zinc oxide nanoparticles (ZnO-NPs) have gained significant attention in cancer therapy due to their unique physical and chemical properties, particularly in treating gastrointestinal (GI) cancers such as gastric, colorectal, and hepatocellular carcinoma. These nanoparticles generate reactive oxygen species (ROS) upon entering cancer cells, causing oxidative stress that leads to cellular damage, DNA fragmentation, and apoptosis. ZnO-NPs affect the expression of key proteins involved in apoptosis, including p53, Bax, and Bcl-2, which regulate cell cycle arrest and programmed cell death. Additionally, ZnO-NPs can reduce mitochondrial membrane potential, further enhancing apoptosis in cancer cells. Furthermore, ZnO-NPs inhibit cancer cell proliferation by interfering with cell cycle progression. They reduce levels of cyclins and cyclin-dependent kinases (CDKs), leading to cell cycle arrest. ZnO-NPs also exhibit anti-metastatic properties by inhibiting the migration and invasion of cancer cells through modulation of signaling pathways that affect cell adhesion and cytoskeletal dynamics. The efficacy of ZnO-NPs in overcoming chemotherapy resistance has been demonstrated by their ability to reduce the IC50 values of chemotherapeutic agents, making cancer cells more susceptible to drug-induced cell death. In this review, we summarize the mechanisms by which ZnO-NPs exert anticancer effects in GI cancers, focusing on apoptosis, cell cycle regulation, and metastasis inhibition, while also highlighting the current limitations in translating these findings into effective clinical treatments.
Collapse
Affiliation(s)
- Yonggang Guo
- Pingdingshan College, Pingdingshan, Henan, China
| | | |
Collapse
|
15
|
Wu Y, Li T, Zhang R, Shi T, Wang S, Zhu L, Zhang Y, Zheng X, Yu X, Zhang J. Establishment of nomogram of early death in elderly pancreatic cancer patients with liver metastasis. Discov Oncol 2025; 16:333. [PMID: 40095230 PMCID: PMC11914455 DOI: 10.1007/s12672-025-02059-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Many elderly patients with pancreatic cancer (PC) often have liver metastasis (LM), and these patients often have poor prognosis and early death (ED). However, few models can accurately predict ED from elderly PC patients with LM. Therefore, we aim to create nomograms to predict ED in elderly PC patients with LM. METHODS All elderly (≥ 60 years old) PC patients with LM from 2010 to 2020 were downloaded from the Surveillance, Epidemiology, and End Result (SEER) database according to the admission criteria. The included data was randomly divided into the training set and the validation set, with a ratio of 7:3. The risk factors for ED in elderly PC patients with LM were determined by univariate and multivariate logistic regression methods, and a nomogram model was established. Lastly, the nomogram is verified by the receiver operating characteristic (ROC) curve, area under the curve (AUC), calibration curve, and decision curve analysis (DCA). RESULTS A total of 1,424 elderly PC patients with LM were randomly divided into training set (n = 996) and validation set (n = 428) based on the ratio of 7:3. The independent prognostic factors for ED include T stage, N stage, surgery, chemotherapy, lung metastasis, and other metastases. These variables were used to create nomograms, where the AUC of the training set and the validation set were 0.83 (95% CI 0.80-0.85) and 0.81 (95% CI 0.77-0.85), respectively. Furthermore, the calibration curve shows that the predicted ED is in good agreement with the actual value. DCA also shows good clinical application value. CONCLUSIONS The developed nomogram can be used to predict the specific probability of ED in elderly PC patients with LM, which is useful in guiding the early prevention and treatment decision-making of this group of people.
Collapse
Affiliation(s)
- Yang Wu
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Tian Li
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
- Department of Nephrology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
| | - Runbing Zhang
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Tingting Shi
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Shunna Wang
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Lingling Zhu
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Yani Zhang
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
- The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Xiaofeng Zheng
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Xiaohui Yu
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China
| | - Jiucong Zhang
- Department of Gastroenterology, The 940th Hospital of Joint Logistic Support Force of PLA, Lanzhou, 730050, Gansu, China.
| |
Collapse
|
16
|
Zhang N, Deng G, Jia R, Han Q, Dai G. Association of lung immune prognostic index with overall survival in pancreatic ductal adenocarcinoma patients treated using chemotherapy. Int J Med Sci 2025; 22:1672-1679. [PMID: 40093811 PMCID: PMC11905270 DOI: 10.7150/ijms.102404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Background: The lung immune prognostic index (LIPI) has attracted considerable interest for its prognostic value in several malignancies. However, its prognostic value in pancreatic ductal adenocarcinoma (PDAC) has not yet been clarified. Objective: This study aimed to assess the role of LIPI with regard to overall survival (OS) in locally advanced or metastatic PDAC patients undergoing chemotherapy. Methods: Data from 256 patients with PDAC treated via chemotherapy at the Chinese PLA General Hospital between January 1, 2011 and July 1, 2018 were retrospectively reviewed. Their neutrophil-to-lymphocyte ratio (dNLR) with lactate dehydrogenase (LDH) values were used to calculate each one's LIPI. The Cox proportional hazard model was used to identify the association between LIPI and OS. Results: Of the included patients, 154 were in the good LIPI group and 102 were in the intermediate/poor LIPI group. The OS in the two groups were 9.0 months (95% CI: 7.351-10.649) and 6.0 months (95% CI: 4.812-7.188), respectively. Patients in the good LIPI group had better OS compared to those in the intermediate/poor LIPI group (HR, 0.720; 95% CI: 0.554-0.935; P = 0.014). Conclusion: This study revealed LIPI is significantly associated with OS in PDAC and could play a significant role in helping clinicians make appropriate decisions for PDAC patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Nan Zhang
- Medical School of Chinese PLA, Beijing 100853, China
| | - Guochao Deng
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing 100071, China
| | - Ru Jia
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing 100071, China
| | - Quanli Han
- Department of Medical Oncology, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Guanghai Dai
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing 100071, China
- Department of Medical Oncology, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
17
|
Karimi E, Kalaki NS, Akrami SM. Identifying Hub Genes and Pathways in Pancreatic Ductal Adenocarcinoma (PAAD): A comprehensive in silico study. Biochem Biophys Rep 2025; 41:101921. [PMID: 39911529 PMCID: PMC11794163 DOI: 10.1016/j.bbrep.2025.101921] [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: 12/10/2024] [Revised: 01/13/2025] [Accepted: 01/13/2025] [Indexed: 02/07/2025] Open
Abstract
Background One of the most aggressive tumors is pancreatic ductal adenocarcinoma (PAAD), which is usually discovered at an advanced stage and is linked to a poor response to current treatment options and a significant risk of metastasis. Methods The Gene Expression Omnibus (GEO) database selected GSE15471, GSE28735, GSE62165, and GSE16515. Differentially expressed genes (DEGs) were defined as having a logFC of >1 and ≤ -1 and an adjusted p-value of less than 0.05. Differentially expressed genes (DEGs) from the four datasets were identified using the GEO2R tool. KEGG and GO databases were used to identify related pathways. PPIs were analyzed using Cytoscape and Gephi. A GEPIA analysis confirmed the target genes. Results The analysis of protein-protein interactions (PPI) along with data from the Gene Expression Omnibus (GEO) led to the identification of 66 hub genes and 819 common differentially expressed genes (DEGs). GO and KEGG pathway analyses indicated that these DEGs are significantly associated with functions related to cell adhesion, extracellular exosomes, structural components of the extracellular matrix, and the cytoskeleton in muscle cells. The expression levels of 8 genes-FN1, CXCR4, MMP9, PXDN, CBS, ALB, GPT2, and EGF-demonstrated a notable difference between normal and tumor samples, as identified through GEPIA analysis. Conclusion The hub genes and related pathways that are connected to the development of PAAD were found in this study. These genes could serve as promising diagnostic biomarkers, offering a valuable chance to detect PAAD in its initial stages, leading to more effective treatment options.
Collapse
Affiliation(s)
- Elham Karimi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloufar Sadat Kalaki
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Seyed Mohammad Akrami
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| |
Collapse
|
18
|
Peng C, Wang J, Wang S, Zhao Y, Wang H, Wang Y, Ma Y, Yang J. Endoplasmic Reticulum Stress: Triggers Microenvironmental Regulation and Drives Tumor Evolution. Cancer Med 2025; 14:e70684. [PMID: 40035165 PMCID: PMC11877002 DOI: 10.1002/cam4.70684] [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: 08/22/2024] [Revised: 12/23/2024] [Accepted: 01/29/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND The endoplasmic reticulum (ER) serves as a crucial hub for protein synthesis and processing, playing an essential role in maintaining protein homeostasis. Perturbations, such as hypoxia, oxidative stress, inadequate amino acid supply, Ca2+ imbalance, and acidosis, can disrupt cellular equilibrium and result in the accumulation of misfolded/unfolded proteins within the ER lumen. This triggers ER stress. In response to this stress, an unfolded protein response (UPR) is activated as a mechanism to cope with the stress and restore internal balance. The UPR is regulated by three sensors located in the ER: inositol-requiring enzyme 1 (IRE1), protein kinase RNA-like endoplasmic reticulum kinase (PERK), and activating transcription factor 6 (ATF6). However, the UPR can promote tumor growth in vivo by affecting tumor angiogenesis, cell migration, cell metabolism, and treatment resistance, and has a huge impact on the tumor microenvironment. MATERIALS AND METHODS We conducted a literature review of scientific papers on the topic of ER stress in the tumor microenvironment. RESULTS AND DISCUSSION This review focuses on the inducing factors of ER stress, the mechanism of the UPR signaling pathway induced by ER stress, and the effect of ER stress on the tumor microenvironment and immune-infiltrating cells. Tumors can regulate their evolution by affecting themselves and the tumor microenvironment through endoplasmic reticulum stress. This study reveals the important role of endoplasmic reticulum stress in the occurrence and development of tumors, and provides new ideas and potential therapeutic targets for the precision treatment of tumors. Future studies can further explore the molecular mechanism of ER stress regulating tumor microenvironment and explore its application potential in clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Chaosheng Peng
- Department of Digestive SurgeryXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
| | - Juan Wang
- Department of Digestive SurgeryXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
| | - Shu Wang
- Department of Digestive SurgeryXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
| | - Yan Zhao
- Department of Digestive SurgeryXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
| | - Haoyuan Wang
- Department of Digestive SurgeryXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
| | - Yuhao Wang
- Department of Digestive SurgeryXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
| | - Yuxuan Ma
- Department of Digestive SurgeryXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive DiseasesXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
| | - Jianjun Yang
- Department of Digestive SurgeryXijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
19
|
Jara CP, Al-Gahmi AM, Lazenby A, Hollingsworth MA, Carlson MA. Selective epithelial expression of KRAS G12D in the Oncopig pancreas drives ductal proliferation and desmoplasia that is accompanied by an immune response. Sci Rep 2025; 15:4736. [PMID: 39922849 PMCID: PMC11807195 DOI: 10.1038/s41598-025-87178-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 01/16/2025] [Indexed: 02/10/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a formidable challenge in oncology, characterized by a high mortality rate, largely attributable to delayed diagnosis and the intricacies of its tumor microenvironment. Innovations in modeling pancreatic epithelial transformation provide valuable insights into the pathogenesis and potential therapeutic strategies for PDAC. We employed a porcine (Oncopig) model, utilizing the Ad-K8-Cre adenoviral vector, to investigate the effects of variable doses (107 to 1010 pfu) on pancreatic epithelial cells. This vector, the expression from which being driven by a Keratin-8 promoter, will deliver Cre-recombinase specifically to epithelial cells. Intraductal pancreatic injections in transgenic Oncopigs (LSL-KRASG12D-TP53R167H) were performed with histologically based evaluation at 2 months post-injection. Specificity of the adenoviral vector was validated through Keratin-8 expression and Cre-recombinase activity. We confirmed that the Ad-K8-Cre adenoviral vector predominantly targets ductal epithelial cells lining both large and small pancreatic ducts, as evidenced by Keratin 8 and CAM5.2 staining. Higher doses resulted in significant tissue morphology changes, including atrophy, and enlarged lymph nodes. Microscopic examination revealed concentration-dependent proliferation of the ductal epithelium, cellular atypia, metaplasia, and stromal alterations. Transgene expression was confirmed with immunohistochemistry. Desmoplastic responses were evident through vimentin, α-SMA, and Masson's trichrome staining, indicating progressive collagen deposition, particularly at the higher vector doses. Our study suggests a distinct dose-response relationship of Ad-K8-Cre in inducing pancreatic epithelial proliferation and possible neoplasia in an Oncopig model. All doses of the vector induced epithelial proliferation; the higher doses also produced stromal alterations, metaplasia, and possible neoplastic transformation. These findings highlight the potential for site-specific activation of oncogenes in large animal models of epithelial tumors, with the ability to induce stromal alterations reminiscent of human PDAC.
Collapse
Affiliation(s)
- Carlos P Jara
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Audrey Lazenby
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael A Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mark A Carlson
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA.
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
20
|
Bonazzi VF, Aoude LG, Brosda S, Bradford JJ, Lonie JM, Loffler KA, Gartside MG, Patel K, Mukhopadhyay P, Keane C, Gebski V, Kench JG, Goldstein D, Waddell N, Barbour AP. C-reactive protein is a prognostic biomarker in pancreatic ductal adenocarcinoma patients. Asia Pac J Clin Oncol 2025; 21:77-86. [PMID: 37415393 PMCID: PMC11733851 DOI: 10.1111/ajco.13993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/04/2023] [Accepted: 06/26/2023] [Indexed: 07/08/2023]
Abstract
AIM The 5-year survival rate of pancreatic ductal adenocarcinoma (PDAC) is approximately 11% and has only improved marginally over the last three decades. For operable PDAC, resection and adjuvant FOLFIRINOX chemotherapy is standard of care. There is growing interest in perioperative regimens to improve outcomes. The non-randomized Phase II study "Gemcitabine and Abraxane for resectable Pancreatic cancer" (GAP) demonstrated the feasibility of perioperative gemcitabine/abraxane. Long-term survival in PDAC requires an effective immune response; hence, we undertook this translational study of the GAP trial cohort to identify immune-oncology biomarkers for clinical use. METHODS We combined Nanostring nCounter technology with immunohistochemistry to investigate the correlation between gene expression and overall patient survival. Findings were investigated in samples from the International Cancer Genome Consortium (ICGC, n = 88) and the Australian Pancreatic Genome Initiative (APGI, n = 227). RESULTS We confirmed that human equilibrative nucleoside transporter 1 (hENT1) expression was not a prognostic marker in PDAC but patients with high levels of hENT1 were more likely to live longer than 24 months post-surgery. Additionally, CD274 (PD-L1) and two novel biomarkers of survival, cathepsin W (CTSW) and C-reactive protein (CRP), were identified in the GAP cohort (n = 19). CRP expression was confirmed in data from the ICGC. Although PD-L1 and CTSW proteins were not significant across all three cohorts, results show that low CRP mRNA and protein expression are associated with longer overall survival in all three patient groups. CONCLUSION PDAC patients with long survival have higher hENT1 expression levels. Furthermore, CRP expression is a biomarker of poor prognosis following perioperative chemotherapy and resection in PDAC patients and thus may be useful for identifying patients who could benefit from more aggressive adjuvant strategies.
Collapse
Affiliation(s)
- Vanessa F. Bonazzi
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Lauren G. Aoude
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Sandra Brosda
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Julia J. Bradford
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - James M. Lonie
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Kelly A. Loffler
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
- College of Medicine and Public HealthFlinders UniversityBedford ParkSouth AustraliaAustralia
| | - Michael G. Gartside
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | - Kalpana Patel
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
| | | | - Colm Keane
- Mater Research Institute‐UQSouth BrisbaneQueenslandAustralia
| | - Val Gebski
- NHMRC Clinical Trials CentreCamperdownNew South WalesAustralia
| | - James G. Kench
- Royal Prince Alfred HospitalCamperdownNew South WalesAustralia
- University of Sydney Central Clinical SchoolCamperdownNew South WalesAustralia
| | - David Goldstein
- University of NSW Prince of Wales Clinical SchoolRandwickNew South WalesAustralia
| | - Nicola Waddell
- QIMRBerghofer Medical Research InstituteHerstonQueenslandAustralia
| | - Andrew P. Barbour
- Frazer InstituteThe University of QueenslandWoolloongabbaQueenslandAustralia
- Princess Alexandra HospitalWoolloongabbaQueenslandAustralia
| | | |
Collapse
|
21
|
Escalera-Anzola S, Rosado M, Yang Y, Parra-Sanchez D, Pedro-Liberal CS, Acedo P. Breakthroughs in nanoparticle-based strategies for pancreatic cancer therapy. Biochem Pharmacol 2025; 232:116685. [PMID: 39613113 DOI: 10.1016/j.bcp.2024.116685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/31/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers worldwide, mainly due to its high heterogeneity, resistance to therapy and late diagnosis, with a 5-year survival rate of less than 10%. This dismal prognosis has promoted strategies to develop more effective treatments. Nanoparticle-based strategies have emerged, in the last decades, as a great opportunity because they can enhance drug delivery and promote controlled release, presenting lower side effects than conventional therapeutic regimens. Moreover, nanoparticles can often be modified to target specific cells or to achieve a sustained release of the drugs into the tumor. However, very few nanoparticle-based therapies are clinically approved. Concretely for pancreatic cancer treatment only two nanoformulations have been approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) so far. Clinical translation of nanoparticles remains a challenge for modern medicine, and in particular for pancreatic cancer therapy, because of the complexity of the disease, and a lack of studies been performed in clinically relevant in vitro and in vivo models. In this review, we have summarized the most recent clinical trials using nanoparticle-based formulations in PDAC, giving a small context of the diverse types of nanoparticles employed and the most recent advancements in the field.
Collapse
Affiliation(s)
- Sara Escalera-Anzola
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom; Smart Devices for Nano Medicine Group, Unidad Excelencia Instituto de BioMedicina y Genética Molecular (IBGM) de Valladolid, University of Valladolid and CSIC, Valladolid, Spain
| | - Maria Rosado
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Yuchen Yang
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Daniel Parra-Sanchez
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Carolina San Pedro-Liberal
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Pilar Acedo
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom.
| |
Collapse
|
22
|
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
|
23
|
Qadir MI, Baril JA, Yip-Schneider MT, Schonlau D, Tran TTT, Schmidt CM, Kolbinger FR. Artificial Intelligence in Pancreatic Intraductal Papillary Mucinous Neoplasm Imaging: A Systematic Review. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.08.25320130. [PMID: 39830259 PMCID: PMC11741484 DOI: 10.1101/2025.01.08.25320130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Background Based on the Fukuoka and Kyoto international consensus guidelines, the current clinical management of intraductal papillary mucinous neoplasm (IPMN) largely depends on imaging features. While these criteria are highly sensitive in detecting high-risk IPMN, they lack specificity, resulting in surgical overtreatment. Artificial Intelligence (AI)-based medical image analysis has the potential to augment the clinical management of IPMNs by improving diagnostic accuracy. Methods Based on a systematic review of the academic literature on AI in IPMN imaging, 1041 publications were identified of which 25 published studies were included in the analysis. The studies were stratified based on prediction target, underlying data type and imaging modality, patient cohort size, and stage of clinical translation and were subsequently analyzed to identify trends and gaps in the field. Results Research on AI in IPMN imaging has been increasing in recent years. The majority of studies utilized CT imaging to train computational models. Most studies presented computational models developed on single-center datasets (n=11,44%) and included less than 250 patients (n=18,72%). Methodologically, convolutional neural network (CNN)-based algorithms were most commonly used. Thematically, most studies reported models augmenting differential diagnosis (n=9,36%) or risk stratification (n=10,40%) rather than IPMN detection (n=5,20%) or IPMN segmentation (n=2,8%). Conclusion This systematic review provides a comprehensive overview of the research landscape of AI in IPMN imaging. Computational models have potential to enhance the accurate and precise stratification of patients with IPMN. Multicenter collaboration and datasets comprising various modalities are necessary to fully utilize this potential, alongside concerted efforts towards clinical translation.
Collapse
Affiliation(s)
| | - Jackson A. Baril
- Division of Surgical Oncology, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michele T. Yip-Schneider
- Division of Surgical Oncology, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Duane Schonlau
- Department of Radiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Thi Thanh Thoa Tran
- Division of Surgical Oncology, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - C. Max Schmidt
- Division of Surgical Oncology, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fiona R. Kolbinger
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
- Regenstrief Center for Healthcare Engineering (RCHE), Purdue University, West Lafayette, IN, USA
- Department of Biostatistics and Health Data Science, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
24
|
Bloemberg J, van Wees S, Kortman VG, Sakes A. Design of a wasp-inspired biopsy needle capable of self-propulsion and friction-based tissue transport. Front Bioeng Biotechnol 2025; 12:1497221. [PMID: 39834634 PMCID: PMC11743259 DOI: 10.3389/fbioe.2024.1497221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/02/2024] [Indexed: 01/22/2025] Open
Abstract
Percutaneous pancreatic core biopsy is conclusive but challenging due to large-diameter needles, while smaller-diameter needles used in aspiration methods suffer from buckling and clogging. Inspired by the ovipositor of parasitic wasps, which resists buckling through self-propulsion and prevents clogging via friction-based transport, research has led to the integration of these functionalities into multi-segment needle designs or tissue transport system designs. This study aimed to combine these wasp-inspired functionalities into a single biopsy needle by changing the interconnection of the needle segments. The resulting biopsy needle features six parallel needle segments interconnected by a ring passing through slots along the length of the needle segments, enabling a wasp-inspired reciprocating motion. Actuation employs a cam and follower mechanism for controlled translation of the segments. The needle prototype, constructed from nitinol rods and stainless steel rings, measures 3 mm in outer diameter and 1 mm in inner diameter. Testing in gelatin phantoms demonstrated efficient gelatin core transport (up to 69.9% ± 9.1% transport efficiency) and self-propulsion (0.842 ± 0.042 slip ratio). Future iterations should aim to reduce the outer diameter while maintaining tissue yield. The design offers a promising new avenue for wasp-inspired medical tools, potentially enhancing early pancreatic cancer detection, thus reducing healthcare costs and patient complications.
Collapse
Affiliation(s)
- Jette Bloemberg
- Department of BioMechanical Engineering, Bio-Inspired Technology Group, Faculty of Mechanical Engineering, Delft University of Technology, Delft, Netherlands
| | - Suzanne van Wees
- Department of BioMechanical Engineering, Bio-Inspired Technology Group, Faculty of Mechanical Engineering, Delft University of Technology, Delft, Netherlands
- Department of Biomedical Engineering, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
| | - Vera G. Kortman
- Department of BioMechanical Engineering, Bio-Inspired Technology Group, Faculty of Mechanical Engineering, Delft University of Technology, Delft, Netherlands
| | - Aimée Sakes
- Department of BioMechanical Engineering, Bio-Inspired Technology Group, Faculty of Mechanical Engineering, Delft University of Technology, Delft, Netherlands
| |
Collapse
|
25
|
Breaza GM, Hut FE, Cretu O, Abu-Awwad SA, Abu-Awwad A, Sima LV, Dan RG, Dan CAM, Closca RM, Zara F. Correlation Between Systemic Inflammation, Gut Microbiome Dysbiosis and Postoperative Complications After the Modified Whipple Procedure. Biomedicines 2025; 13:104. [PMID: 39857688 PMCID: PMC11762099 DOI: 10.3390/biomedicines13010104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
(1) Background: The modified Whipple procedure, or pylorus-preserving pancreaticoduodenectomy, is a complex surgical intervention used to treat pancreatic head tumors. While preserving digestive function, it is associated with significant perioperative risks. This study explores the clinical, immunological, and microbiome-related factors influencing postoperative complications, focusing on the interplay between patient comorbidities, systemic inflammation, and gut dysbiosis. (2) Methods: A retrospective analysis was conducted on 123 patients undergoing the modified Whipple procedure for pancreatic head tumors. Patients were categorized into two groups based on the occurrence of significant postoperative complications (Group A: with complications; Group B: without complications). Data on demographics, comorbidities, inflammatory markers (CRP, IL-6, procalcitonin), and gut microbiome composition were collected. Microbial diversity was evaluated using the Shannon Index, and logistic regression was performed to identify independent predictors of complications. (3) Results: Patients in Group A had a significantly higher prevalence of diabetes mellitus (43.1% vs. 20.8%; p = 0.02) and cardiovascular disease (35.3% vs. 13.9%; p = 0.01). Elevated inflammatory markers (CRP ≥ 40 mg/L, IL-6 ≥ 30 pg/mL, procalcitonin ≥ 0.5 ng/mL) were strongly associated with higher complication rates. Microbiome analysis indicated dysbiosis in Group A, with reduced Lactobacillus and Bifidobacterium levels, increased Enterobacteriaceae abundance, and a lower Shannon Index (<2). Patients exhibiting both dysbiosis and elevated inflammation had the highest complication rate (60%). Multivariate analysis identified diabetes, elevated IL-6, and dysbiosis as independent predictors of adverse outcomes. (4) Conclusions: Postoperative complications after the modified Whipple procedure are influenced by systemic inflammation and gut dysbiosis. A systematic preoperative assessment of microbiome health and inflammatory markers enables accurate risk stratification and personalized interventions, potentially reducing the incidence of complications and improving overall surgical outcomes.
Collapse
Affiliation(s)
- Gelu Mihai Breaza
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- University Clinic of Surgery I, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (O.C.); (L.V.S.); (R.G.D.); (C.A.-M.D.)
| | - Florin Emil Hut
- University Clinic of Surgery I, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (O.C.); (L.V.S.); (R.G.D.); (C.A.-M.D.)
- Center for Hepato-Bilio-Pancreatic Surgery, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Octavian Cretu
- University Clinic of Surgery I, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (O.C.); (L.V.S.); (R.G.D.); (C.A.-M.D.)
| | - Simona-Alina Abu-Awwad
- Ist Clinic of Obstetrics and Gynecology, “Pius Brinzeu” County Clinical Emergency Hospital, 300723 Timisoara, Romania;
- Department of Obstetrics and Gynecology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Ahmed Abu-Awwad
- Department XV—Discipline of Orthopedics—Traumatology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Research Center University Professor Doctor Teodor Sora, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Laurențiu Vasile Sima
- University Clinic of Surgery I, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (O.C.); (L.V.S.); (R.G.D.); (C.A.-M.D.)
| | - Radu Gheorghe Dan
- University Clinic of Surgery I, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (O.C.); (L.V.S.); (R.G.D.); (C.A.-M.D.)
| | - Cristina Ana-Maria Dan
- University Clinic of Surgery I, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (O.C.); (L.V.S.); (R.G.D.); (C.A.-M.D.)
| | - Raluca Maria Closca
- Department of Pathology, Emergency City Hospital, 300254 Timisoara, Romania; (R.M.C.); (F.Z.)
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania
| | - Flavia Zara
- Department of Pathology, Emergency City Hospital, 300254 Timisoara, Romania; (R.M.C.); (F.Z.)
- Department of Microscopic Morphology, University of Medicine and Pharmacy “Victor Babes”, 300041 Timisoara, Romania
| |
Collapse
|
26
|
Rupareliya M, Shende P. Influence of RNA Methylation on Cancerous Cells: A Prospective Approach for Alteration of In Vivo Cellular Composition. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1474:79-103. [PMID: 39259424 DOI: 10.1007/5584_2024_820] [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: 09/13/2024]
Abstract
RNA methylation is a dynamic and ubiquitous post-transcriptional modification that plays a pivotal role in regulating gene expression in various conditions like cancer, neurological disorders, cardiovascular diseases, viral infections, metabolic disorders, and autoimmune diseases. RNA methylation manifests across diverse RNA species including messenger RNA (mRNA), ribosomal RNA (rRNA), and transfer RNA (tRNA), exerting pivotal roles in gene expression regulation and various biological phenomena. Aberrant activity of writer, eraser, and reader proteins enables dysregulated methylation landscape across diverse malignancy transcriptomes, frequently promoting cancer pathogenesis. Numerous oncogenic drivers, tumour suppressors, invasion/metastasis factors, and signalling cascade components undergo methylation changes that modulate respective mRNA stability, translation, splicing, transport, and protein-RNA interactions accordingly. Functional studies confirm methylation-dependent alterations drive proliferation, survival, motility, angiogenesis, stemness, metabolism, and therapeutic evasion programs systemically. Methyltransferase overexpression typifies certain breast, liver, gastric, and other carcinomas correlating with adverse clinical outcomes like diminished overall survival. Mapping efforts uncover nodal transcripts for targeted drug development against hyperactivated regulators including METTL3. Some erasers and readers also suitable lead candidates based on apparent synthetic lethality. Proteomic screens additionally highlight relevant methylation-sensitive effector pathways amenable to combinatorial blockade, reversing compensatory signalling mechanisms that facilitate solid tumour progression. Quantifying global methylation burdens and responsible enzymes clinically predicts patient prognosis, risk stratification for adjuvant therapy, and overall therapeutic responsiveness.
Collapse
Affiliation(s)
- Manali Rupareliya
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, India
| | - Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, India.
| |
Collapse
|
27
|
Stefàno E, Rovito G, Cossa LG, Castro FD, Vergaro V, Ali A, My G, Migoni D, Muscella A, Marsigliante S, Benedetti M, Fanizzi FP. Novel Pt (II) Complexes With Anticancer Activity Against Pancreatic Ductal Adenocarcinoma Cells. Bioinorg Chem Appl 2024; 2024:5588491. [PMID: 39886428 PMCID: PMC11779987 DOI: 10.1155/bca/5588491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/16/2024] [Indexed: 02/01/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive type of solid tumor that is becoming more common. cis-[PtCl2 (NH3)2] (in short cisplatin or CDDP) has been shown to be effective in treating various cancers, including PDAC. However, the development of resistance to chemotherapy drugs has created a need for the synthesis of new anticancer agents. Platinum-based drugs containing the bidentate ligand phenanthroline have been found to have strong antitumor activity due to their ability to cause DNA damage. In this study, we examined the ability of two Pt (II) cationic complexes, [Pt(η 1-C2H4OR) (DMSO) (phen)]+ (in short Pt-EtORSOphen; R = Me, 1; Et, 2), to inhibit the growth and spread of BxPC-3 PDAC cells, in comparison to CDDP. The length of the alkyl chain and its associated lipophilic properties did not affect the anticancer effects of complexes 1 and 2 in BxPC-3 cells. However, it did appear to influence the rapid loss of mitochondrial membrane potential (ΔΨM), suggesting that these complexes could potentially be used as mitochondria-targeted lipophilic cations in anticancer therapy.
Collapse
Affiliation(s)
- Erika Stefàno
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Gianluca Rovito
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Luca G. Cossa
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Federica De Castro
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Viviana Vergaro
- Department of Experimental Medicine, University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Asjad Ali
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Giulia My
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Danilo Migoni
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Antonella Muscella
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Santo Marsigliante
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Michele Benedetti
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| | - Francesco Paolo Fanizzi
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Via Monteroni I-73100, Italy
| |
Collapse
|
28
|
Jin Y, Hu R, Gu Y, Wei A, Li A, Zhang Y. Quantitative site-specific N-glycosylation analysis reveals IgG glyco-signatures for pancreatic cancer diagnosis. Clin Proteomics 2024; 21:68. [PMID: 39734184 DOI: 10.1186/s12014-024-09522-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024] Open
Abstract
BACKGROUND Pancreatic cancer is a highly aggressive tumor with a poor prognosis due to a low early detection rate and a lack of biomarkers. Most of pancreatic cancer is pancreatic ductal adenocarcinoma (PDAC). Alterations in the N-glycosylation of plasma immunoglobulin G (IgG) have been shown to be closely associated with the onset and development of several cancers and could be used as biomarkers for diagnosis. The study aimed to explore intact N-glycosylation profile of IgG in patients with PDAC and find relation between intact N-glycosylation profile of IgG and clinical information such as diagnosis and prognosis. METHODS In this study, we employed a well-evaluated approach (termed GlycoQuant) to assess the site-specific N-glycosylation profile of human plasma IgG in both healthy individuals and patients with pancreatic ductal adenocarcinoma (PDAC). The datasets generated and analyzed during the current study are available in the ProteomeXchange Consortium ( http://www.proteomexchange.org/ ) via the iProX partner repository, with the dataset identifier PXD051436. RESULTS The analysis of rapidly purified IgG samples from 100 patients with different stages of PDAC, in addition to 30 healthy controls, revealed that the combination of carbohydrate antigen 19 - 9 (CA19-9), IgG1-GP05 (IgG1-TKPREEQYNSTYR-HexNAc [4]Hex [5]Fuc [1]NeuAc [1]), and IgG4-GP04 (IgG4-EEQFNSTYR- HexNAc [4]Hex [5]Fuc [1]NeuAc [1]) can be used to distinguish between PDAC patients and healthy individuals (AUC = 0.988). In addition, cross validation of the diagnosis model showed satisfactory result. CONCLUSIONS The study demonstrated that the integrated quantitative method can be utilized for large-scale clinical N-glycosylation research to identify novel N-glycosylated biomarkers. This could facilitate the development of clinical glycoproteomics.
Collapse
Affiliation(s)
- Yi Jin
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan, 610041, China
| | - Ran Hu
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan, 610041, China
| | - Yufan Gu
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, Keyuan 4th Road, Gaopeng Avenue, Hi-tech Zone, Chengdu, Sichuan, 610041, China
| | - Ailin Wei
- Guang'an People's Hospital, Guang'an, 638001, China
| | - Ang Li
- Guang'an People's Hospital, Guang'an, 638001, China.
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan, 610041, China.
| | - Yong Zhang
- Department of Pancreatic Surgery and Institutes for Systems Genetics, West China Hospital, Sichuan University, Keyuan 4th Road, Gaopeng Avenue, Hi-tech Zone, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
29
|
Bhatt P, Hirsch J, Cockrum P, Kim G, Dieguez G. The Effects of Adverse Events and Associated Costs on Value-Based Care for Metastatic Pancreatic Ductal Adenocarcinoma. JOURNAL OF HEALTH ECONOMICS AND OUTCOMES RESEARCH 2024; 11:161-167. [PMID: 39717859 PMCID: PMC11664866 DOI: 10.36469/001c.124367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/03/2024] [Indexed: 12/25/2024]
Abstract
Background: Rising oncology healthcare costs have led to value-based care reimbursement models that coordinate care and improve quality while reducing overall spending. These models are increasingly important for traditional Medicare and other payers. Objectives: To compare the incidence of adverse events (AEs), AE-associated excess costs, and total cost of care (TCOC) of 3 cohorts receiving first-line treatment for metastatic pancreatic ductal adenocarcinoma (mPDAC). Methods: We conducted a retrospective analysis of administrative claims data from 2018 to 2022 using the Medicare 100% Research Identifiable Files. We examined 3 cohorts receiving mPDAC treatment: FOLFIRINOX (FFX) (oxaliplatin, irinotecan, leucovorin, 5-FU bolus and infusion); modified FFX, (5-FU infusion only); and gemcitabine/nab-paclitaxel (gem/abrax). We compared the incidence of clinically significant AEs, TCOC, components of TCOC, and costs related to AEs/treatment toxicity. Results: Patient AE rates ranged from 6.2% to 51.7%. AEs occurred more frequently in patients receiving FFX with all 4 components. Patients receiving brand name gem/abrax had lower rates of febrile neutropenia (6.2%) and neutropenia (22.2%) than those receiving FFX with no 5-FU bolus (febrile neutropenia, 9.9%; neutropenia, 36.9%) and FFX with all 4 components (febrile neutropenia, 6.9%; neutropenia, 30.4%). Rates of most nonhematologic AEs were higher in patients receiving FFX with all 4 components, with diarrhea occurring in 28.3%, abdominal pain in 31.5%, and nausea/vomiting in 41.5% of patients. TCOC was lower in the gem/abrax cohort: 6505 v s F F X w i t h n o 5 - F U b o l u s ( 6995) and FFX with all 4 components ( 7142 ) p e r a d m i n i s t r a t i o n . T h e d e v e l o p m e n t o f a n y s t u d i e d h e m a t o l o g i c A E w a s a s s o c i a t e d w i t h a m e a n e x c e s s c o s t o f 5993 per administration, while the development of any studied nonhematological AE was associated with a mean per-administration excess cost of $3665. Discussion: Treatment decisions intended to minimize chemotherapy costs may lead to suboptimal decisions if the goal is to reduce TCOC. Our research suggests FFX is more costly than gem/abrax (TCOC per administration). Patients receiving gem/abrax were older and had higher baseline Charlson Comorbidity Index scores; however, other factors may be important in driving cost differences. Conclusions: Irrespective of drug cost, chemotherapy leading to a significant increase in AEs is associated with higher TCOC.
Collapse
|
30
|
Balakrishnan K, Xiao Y, Chen Y, Dong J. Elevated Expression of Cell Adhesion, Metabolic, and Mucus Secretion Gene Clusters Associated with Tumorigenesis, Metastasis, and Poor Survival in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:4049. [PMID: 39682235 DOI: 10.3390/cancers16234049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
OBJECTIVES Technological advances in identifying gene expression profiles are being applied to study an array of cancers. The goal of this study was to identify differentially expressed genes in pancreatic ductal adenocarcinoma (PDAC) and examine their potential role in tumorigenesis and metastasis. METHODS The transcriptomic profiles of PDAC and non-tumorous tissue samples were derived from the gene expression omnibus (GEO), which is a public repository. The GEO2R tool was used to further derive differentially expressed genes from those profiles. RESULTS In this study, a total of 68 genes were derived from upregulated PDAC genes in three or more transcriptomic profiles and were considered PDAC gene sets. The identified PDAC gene sets were examined in the molecular signatures database (MSigDB) for ontological investigation, which revealed that these genes were involved in the extracellular matrix and associated with the cell adhesion process in PDAC tumorigenesis. The gene set enrichment analysis showed greater enrichment scores for the gene sets. Moreover, the identified gene sets were examined for protein-protein interaction using the STRING database. Based on functional k-means clustering, the following three functional cluster groups were identified in this study: extracellular matrix/cell adhesion, metabolic, and mucus secretion-related protein groups. The receiver operating characteristic (ROC) curve revealed greater specificity and sensitivity for these cluster genes in predicting PDAC tumorigenesis and metastases. In addition, the expression of the cluster genes affects the overall survival rate of PDAC patients. Using the cancer genome atlas (TCGA) database, the associations between expression levels and clinicopathological features were validated. CONCLUSIONS Overall, the genes identified in this study appear to be critical in PDAC development and can serve as potential diagnostic and prognostic targets for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Karthik Balakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yi Xiao
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yuanhong Chen
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jixin Dong
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
31
|
Mortoglou M, Lian M, Miralles F, Dart DA, Uysal-Onganer P. miR-210 Mediated Hypoxic Responses in Pancreatic Ductal Adenocarcinoma. ACS OMEGA 2024; 9:47872-47883. [PMID: 39651070 PMCID: PMC11618397 DOI: 10.1021/acsomega.4c08947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 12/11/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one among the most lethal malignancies due to its aggressive behavior and resistance to conventional therapies. Hypoxia significantly contributes to cancer progression and therapeutic resistance of PDAC. microRNAs (miRNAs/miRs) have emerged as critical regulators of various biological processes. miR-210 is known as the "hypoxamir" due to its prominent role in cellular responses to hypoxia. In this study, we investigated the multifaceted role of miR-210 in PDAC using miR-210 knockout (KO) cellular models to elucidate its functions under hypoxic conditions. Hypoxia-inducible factor-1α (HIF1-α), a key transcription factor activated in response to low oxygen levels, upregulates miR-210. miR-210 maintains cancer stem cell (CSC) phenotypes and promotes epithelial-mesenchymal transition (EMT), which is essential for tumor initiation, metastasis, and therapeutic resistance. Our findings demonstrate that miR-210 regulates the expression of CSC markers, such as CD24, CD44, and CD133, and EMT markers, including E-cadherin, Vimentin, and Snail. Specifically, depletion of miR-210 reversed EMT and CSC marker expression levels in hypoxic Panc-1 and MiaPaCa-2 PDAC cells. These regulatory actions facilitate a more invasive and treatment-resistant PDAC phenotype. Understanding the regulatory network involving miR-210 under hypoxic conditions may reveal new therapeutic targets for combating PDAC and improving patient outcomes. Our data suggest that miR-210 is a critical regulator of HIF1-α expression, EMT, and the stemness of PDAC cells in hypoxic environments.
Collapse
Affiliation(s)
- Maria Mortoglou
- Cancer
Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, U.K.
| | - Mutian Lian
- Cancer
Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, U.K.
| | - Francesc Miralles
- School
of Health and Medical Sciences, City St
George’s, University of London, Cranmer Terrace, London SW17 0RE, U.K.
| | - D. Alwyn Dart
- UCL
Cancer Institute, University College London, Paul O’Gorman Building, 72
Huntley Street, London WC1E 6DD, U.K.
| | - Pinar Uysal-Onganer
- Cancer
Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, U.K.
| |
Collapse
|
32
|
Costa VDCMD, Lima MDCAD, da Cruz Filho IJ, Galdino LV, Pereira MC, Silva BDO, Albuquerque APDB, da Rosa MM, Pitta MGDR, Rêgo MJBDM. 5-nitro-thiophene-thiosemicarbazone derivative induces cell death, cell cycle arrest, and phospho-kinase shutdown in pancreatic ductal adenocarcinoma cells. Eur J Pharmacol 2024; 983:176963. [PMID: 39260813 DOI: 10.1016/j.ejphar.2024.176963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/08/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with limited treatment options. This study explores the potential of novel 5-nitro-thiophene-thiosemicarbazone derivatives as therapeutic agents for PDAC. METHODS We evaluated the cytotoxicity of seven derivatives in peripheral blood mononuclear cells (PBMCs) and PDAC cell lines. Promising candidates (PR12 and PR17) were further analyzed for their effects on colony formation, cell cycle progression, and reactive oxygen species (ROS) production. PR17, the most promising derivative, was subjected to additional investigation, including analysis of autophagy-related genes and protein kinase inhibition. RESULTS Three derivatives (PR16, PR19, and PR20) displayed cytotoxicity towards PBMCs. PR12 reduced colony formation and G0/G1 cell cycle arrest in PDAC cells. Notably, PR17 exhibited potent activity in MIA PaCa-2 cells, inducing S-phase cell cycle arrest, downregulating autophagy genes, and inhibiting key protein kinases. CONCLUSION PR17, a 5-nitro-thiophene-thiosemicarbazone derivative, demonstrates promising antineoplastic activity against PDAC cells by potentially modulating cell cycle progression, autophagy, and protein kinase signaling. Further studies are warranted to elucidate the detailed mechanism of action and explore its efficacy in vivo.
Collapse
Affiliation(s)
| | | | | | - Lília Vieira Galdino
- Therapeutic Innovation Research Center - Suely Galdino, Federal University of Pernambuco, Pernambuco, Recife, Brazil
| | - Michelly Cristiny Pereira
- Therapeutic Innovation Research Center - Suely Galdino, Federal University of Pernambuco, Pernambuco, Recife, Brazil
| | - Bárbara de Oliveira Silva
- Therapeutic Innovation Research Center - Suely Galdino, Federal University of Pernambuco, Pernambuco, Recife, Brazil
| | | | - Michelle Melgarejo da Rosa
- Therapeutic Innovation Research Center - Suely Galdino, Federal University of Pernambuco, Pernambuco, Recife, Brazil
| | - Maira Galdino da Rocha Pitta
- Therapeutic Innovation Research Center - Suely Galdino, Federal University of Pernambuco, Pernambuco, Recife, Brazil
| | | |
Collapse
|
33
|
Jiang L, Lu X, Dai Y, Jiang K, Miao Y, Yu J, Yin L, Wei J. Establishment and analysis of a prognostic model of pancreatic ductal adenocarcinomas based on nerve-cancer crosstalk-related genes. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2024; 17:396-410. [PMID: 39660330 PMCID: PMC11626290 DOI: 10.62347/ghum8504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 10/26/2024] [Indexed: 12/12/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor with a five-year survival rate of 13%, the lowest among all malignant tumors. The work aims to use bioinformatics methods to mine Nerve-cancer crosstalk-related genes (NCCGs) in pancreatic cancer and evaluate their correlation with tumor stage and prognosis, thereby providing a new direction of development and experimental basis for pancreatic cancer treatment. This study included 185 individuals with PDAC from the TCGA database, together with clinical and RNA sequencing data. A review of prior studies revealed the mechanism of neural-cancer crosstalk and identified 42 neural-cancer crosstalk-related genes (NCCGs). Multivariate logistic regression analysis showed that NGFR (OR=39.076, 95% CI; P<0.05), CHRNB2 (OR=41.076, 95% CI; P<0.05), and CHRNA10 (OR=39.038, 95% CI; P<0.05) were identified as independent risk factors for PNI development. Pearson correlation analysis revealed that CHRNA10 was negatively connected with PDAC microsatellite instability, whereas CHRNA10, CHRNB2, and NGFR were negatively correlated with PDAC tumor mutation burden. The GEPIA database revealed that CHRNB2 expression was higher in stage I PDAC. The pancreatic cancer single-cell dataset PAAD_CRA001160 revealed that malignant tumor cells, ductal cells, endothelial cells and fibroblasts accounted for a large proportion in the tumor microenvironment of pancreatic cancer. Furthermore, the NGFR gene was shown to be more significantly expressed in various pancreatic cancer cells. Bioinformatics analysis was used to create a validated prognostic model of pancreatic cancer, which explored the critical mechanisms of neural-tumor interactions and revealed the potential of cancer-neural crosstalk-related genes as prognostic biomarkers and anti-tumor therapy targets.
Collapse
Affiliation(s)
- Lei Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Xiaozhi Lu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Yuran Dai
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Kuirong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Yi Miao
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Jun Yu
- Department of Surgery, Johns Hopkins University School of MedicineBaltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence, Johns Hopkins University School of MedicineBaltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Lingdi Yin
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| | - Jishu Wei
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical UniversityNanjing, Jiangsu, China
- Pancreas Institute of Nanjing Medical UniversityNanjing, Jiangsu, China
| |
Collapse
|
34
|
Anastasiou IA, Sarantis P, Rebelos E, Eleftheriadou I, Tentolouris KN, Katsaouni A, Koustas E, Kokala V, Karamouzis MV, Tentolouris N. l-Securinine Induces ROS-Dependent Apoptosis on Pancreatic Cancer Cells via the PI3K/AKT/mTOR Signaling Pathway. J Biochem Mol Toxicol 2024; 38:e70036. [PMID: 39467148 DOI: 10.1002/jbt.70036] [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: 04/30/2024] [Revised: 09/03/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
Accumulating evidence has shown that l-securinine can, in certain circumstances, suppress tumor development by elevating reactive oxygen species (ROS) levels. The current work set out to examine l-securinine's apoptotic effects on HuP-T3 cells as well as any potential underlying molecular mechanism(s) that could explain its action as an anticancer agent. In this study, we used 1.2B4 cells as a control human cell line to verify our findings. Hup-T3 and 1.2B4 cells were cultured with a medium containing the following dilutions of l-securinine: 1-10 μΜ for up to 72 h. We examined the viability and proliferation levels of cells in both cell lines. Then, we measured only 1.2B4 insulin levels and content. We also quantified cell apoptosis, cell cycle levels, and the intracellular reactive oxygen species on HuP-T3 and 1.2B4. Afterwards, we performed a real-time quantitative polymerase chain reaction and western blot analysis. Our results demonstrated that l-securinine inhibited both proliferation and growth of Hup-T3 cells, showing inhibitory and antiproliferative activity in comparison with the control group. In addition, l-securinine had no impact on the proliferation and growth of 1.2B4 cells, nor on their insulin levels and content. By boosting ROS production, and inhibiting the PI3K/AKT/mTOR signaling pathway, l-securinine induced apoptosis on HuP-T3 cells. Pancreatic cancer was successfully inhibited by l-securinine in vitro. l-securinine triggers ROS-dependent apoptosis on pancreatic cancer cells while inhibiting the PI3K/AKT/mTOR signaling pathway. These findings suggest that l-securinine holds promise as a potential lead for future drug development in the fight against pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Ioanna A Anastasiou
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Greece
| | - Eleni Rebelos
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
- Turku PET Centre, University of Turku, Turku, Finland
| | - Ioanna Eleftheriadou
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Konstantinos N Tentolouris
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasia Katsaouni
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Koustas
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Greece
| | - Vasileia Kokala
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Greece
| | - Nikolaos Tentolouris
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| |
Collapse
|
35
|
Aziz H, Kwon YIC, Park AMG, Lai A, Lee KYC, Zhang D, Kwon Y, Pawlik TM. Recent advancements in management for noncolorectal, nonneuroendocrine hepatic metastases. J Gastrointest Surg 2024; 28:1922-1932. [PMID: 39154708 DOI: 10.1016/j.gassur.2024.08.012] [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: 07/29/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Owing to the heterogeneity of underlying primary tumors, noncolorectal, nonneuroendocrine metastases to the liver (NCNNMLs), although relatively rare, pose major challenges to treatment and long-term management. Despite being considered the gold standard for colorectal cancer liver metastases, the role of surgical resection for NCNNML remains controversial. Furthermore, advancements in locoregional treatment modalities, such as ablation and various chemotherapeutic modalities, have contributed to the treatment of patients with NCNNML. METHODS This was a comprehensive review of literature that used Medline/PubMed, Google Scholar, the Cochrane Library, and the Web of Science, which were accessed between 2014 and 2024. RESULTS NCNNMLs are rare tumor entities with varied presentation and outcomes. A multidisciplinary approach, which includes chemotherapy, surgery, and interventional radiologic techniques, can be implemented with good results. CONCLUSION Given the complex nature of NCNNML, its management should be highly individualized and multidisciplinary. Locoregional treatments, such as surgical resection and/or ablation, may be more appropriate for select patients and should be offered as a viable therapeutic option for a subset of individuals.
Collapse
Affiliation(s)
- Hassan Aziz
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Ye In Christopher Kwon
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Andrew Min-Gi Park
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Alan Lai
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Kerry Yi Chen Lee
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Dean Zhang
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Yeseo Kwon
- Department of Surgery, School of Medicine, Tufts University, Boston, MA, United States
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States.
| |
Collapse
|
36
|
Dobos NK, Garay T, Herold M, Simon A, Madar-Dank V, Balka G, Gajdacsi J, Dank M, Szasz AM, Herold Z. Immune Marker and C-Reactive Protein Dynamics and Their Prognostic Implications in Modulated Electro-Hyperthermia Treatment in Advanced Pancreatic Cancer: A Retrospective Analysis. IMMUNO 2024; 4:385-399. [DOI: 10.3390/immuno4040025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
Background: Previous research has suggested that modulated electro-hyperthermia (mEHT) can be used to induce anti-tumor immune effects and to extend patient survival. The use of mEHT in advanced pancreatic cancer is beneficial; however, its immune-mediating effects were never investigated. Methods: A retrospective observational study was conducted. Leukocyte counts, C-reactive protein (CRP), neutrophil-to-lymphocyte ratio (NLR), and granulocyte-to-lymphocyte ratio (GLR) were measured at baseline, midpoint, and after mEHT treatment. Results: A total of 73 mEHT treated pancreatic cancer patients were included. The time elapsed between tumor diagnosis and the first mEHT treatment was 4.40 ± 5.70 months. While no change could be observed between the baseline and the first follow-up visits, the total white blood cell (WBC), neutrophil, and granulocyte count, CRP, NLR, and GLR were significantly higher at the second follow-up compared to both previous visits. Higher levels of the latter parameters following the last mEHT treatment were signaling significantly poor prognostic signs, and so were their longitudinal changes. Conclusions: After the initiation of mEHT, immune markers stabilize with the treatment, but this positive effect is eroded over time by progressive disease. Monitoring the changes in these markers and the occurrence of their increase is a prognostic marker of shorter survival.
Collapse
Affiliation(s)
- Nikolett Kitti Dobos
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary
| | - Tamas Garay
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Magdolna Herold
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
- Department of Internal Medicine and Hematology, Semmelweis University, H-1088 Budapest, Hungary
| | - Alexandra Simon
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary
| | | | - Gyula Balka
- Department of Pathology, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
| | - Jozsef Gajdacsi
- Clinical Center, Semmelweis University, H-1083 Budapest, Hungary
| | - Magdolna Dank
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Attila Marcell Szasz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| | - Zoltan Herold
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
| |
Collapse
|
37
|
Rodriguez VI, Mammadova J, Permuth JB, Luthra A, Pena L, Friedman M, Dam A, Cappelle S, Malafa MP, Hallmon C, Miranda C, Mok SRS. Elevated Urinary Levels of Fungal and Environmental Toxins in Patients with Pancreatic Ductal Adenocarcinoma. J Gastrointest Cancer 2024; 56:4. [PMID: 39419859 PMCID: PMC11486816 DOI: 10.1007/s12029-024-01125-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Risk factors for pancreatic ductal adenocarcinoma (PDAC) include tobacco/alcohol abuse, genetic predisposition, insulin resistance, and pancreatic cysts. Despite these well-established risk factors and the screening of high-risk individuals, some people still develop PDAC. This study aims to explore a potential risk factor for PDAC by investigating the association between fungal toxins (FT) and environmental toxins (ET) and the disease. We predicted that individuals with PDAC would have higher levels of these toxins compared to healthy controls. The rationale behind this hypothesis is that exposure to FT and ET might contribute to the development of PDAC by elevating cancer risk. METHODS A pilot retrospective cohort study was conducted at Moffitt Cancer Center from 2022 to 2023. This study compared FT and ET levels, demographic data, and PDAC features between subjects with PDAC and healthy controls. RESULTS Forty subjects were enrolled in the study, comprising 20 with pancreatic ductal adenocarcinoma (PDAC) and 20 healthy controls. Baseline demographics were similar between the two groups. Among the PDAC subjects, the most common tumor location was the head of the pancreas (55%); 30% had locally advanced disease, 45% were borderline resectable, and 10% had metastatic disease. Compared to the controls, subjects with PDAC had significantly higher levels of fungal toxins (FTs) including ochratoxin, gliotoxin, and citrinin (p < 0.05). Additionally, PDAC patients had significantly elevated levels of environmental toxins (ETs) such as methyl tert-butyl ether (MTBE), xylene, styrene, acrylonitrile, perchlorate, diphenyl phosphate, bromopropane, organophosphates, acrolein, tiglylglycine, and diethylphosphate (p < 0.05). CONCLUSION Our study demonstrates that subjects with PDAC, without other risk factors, have higher FT and ET levels than controls. Further studies are needed to evaluate whether ET and FT exposure can be clinically utilized as a risk factor for PDAC development.
Collapse
Affiliation(s)
- Vanessa I Rodriguez
- Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Jamila Mammadova
- Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jennifer B Permuth
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Anjuli Luthra
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Luis Pena
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Mark Friedman
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Aamir Dam
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Saraswathi Cappelle
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Mokenge P Malafa
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Candice Hallmon
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Cassandra Miranda
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Shaffer R S Mok
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
38
|
Donahue KL, Watkoske HR, Kadiyala P, Du W, Brown K, Scales MK, Elhossiny AM, Espinoza CE, Lasse Opsahl EL, Griffith BD, Wen Y, Sun L, Velez-Delgado A, Renollet NM, Morales J, Nedzesky NM, Baliira RK, Menjivar RE, Medina-Cabrera PI, Rao A, Allen B, Shi J, Frankel TL, Carpenter ES, Bednar F, Zhang Y, Pasca di Magliano M. Oncogenic KRAS-Dependent Stromal Interleukin-33 Directs the Pancreatic Microenvironment to Promote Tumor Growth. Cancer Discov 2024; 14:1964-1989. [PMID: 38958646 PMCID: PMC11450371 DOI: 10.1158/2159-8290.cd-24-0100] [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: 01/30/2024] [Revised: 05/18/2024] [Accepted: 07/01/2024] [Indexed: 07/04/2024]
Abstract
Pancreatic cancer is characterized by an extensive fibroinflammatory microenvironment. During carcinogenesis, normal stromal cells are converted to cytokine-high cancer-associated fibroblasts (CAF). The mechanisms underlying this conversion, including the regulation and function of fibroblast-derived cytokines, are poorly understood. Thus, efforts to therapeutically target CAFs have so far failed. Herein, we show that signals from epithelial cells expressing oncogenic KRAS-a hallmark pancreatic cancer mutation-activate fibroblast autocrine signaling, which drives the expression of the cytokine IL33. Stromal IL33 expression remains high and dependent on epithelial KRAS throughout carcinogenesis; in turn, environmental stress induces interleukin-33 (IL33) secretion. Using compartment-specific IL33 knockout mice, we observed that lack of stromal IL33 leads to profound reprogramming of multiple components of the pancreatic tumor microenvironment, including CAFs, myeloid cells, and lymphocytes. Notably, loss of stromal IL33 leads to an increase in CD8+ T-cell infiltration and activation and, ultimately, reduced tumor growth. Significance: This study provides new insights into the mechanisms underlying the programming of CAFs and shows that during this process, expression of the cytokine IL33 is induced. CAF-derived IL33 has pleiotropic effects on the tumor microenvironment, supporting its potential as a therapeutic target.
Collapse
Affiliation(s)
| | - Hannah R. Watkoske
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, Michigan.
| | - Padma Kadiyala
- Immunology Graduate Program, University of Michigan, Ann Arbor, Michigan.
| | - Wenting Du
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
| | - Kristee Brown
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
| | - Michael K. Scales
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
| | - Ahmed M. Elhossiny
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan.
| | | | | | | | - Yukang Wen
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
| | - Lei Sun
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
| | - Ashley Velez-Delgado
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
| | - Nur M. Renollet
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, Michigan.
| | - Jacqueline Morales
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
| | - Nicholas M. Nedzesky
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, Michigan.
| | | | - Rosa E. Menjivar
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan.
| | | | - Arvind Rao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
- Cancer Data Science Resource, University of Michigan, Ann Arbor, Michigan.
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan.
| | - Benjamin Allen
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| | - Jiaqi Shi
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
- Department of Pathology and Clinical Labs, University of Michigan, Ann Arbor, Michigan.
| | - Timothy L. Frankel
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| | - Eileen S. Carpenter
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Filip Bednar
- Cancer Biology Program, University of Michigan, Ann Arbor, Michigan.
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| | - Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
39
|
Silli EK, Zheng Z, Zhou X, Li M, Tang J, Guo R, Tan C, Wang Y. Design optimization of Fucoidan-coating Cationic Liposomes for enhance Gemcitabine delivery. Invest New Drugs 2024; 42:518-530. [PMID: 39154300 DOI: 10.1007/s10637-024-01455-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/26/2024] [Indexed: 08/19/2024]
Abstract
Obstacles facing chemotherapeutic drugs for cancers led scientists to load Gemcitabine (GEM) into nanocarriers like liposomes, known for their nontoxicity profile and targeting capacity. The liposomal nanostructures containing GEM were coated with Fucoidan (FU) due to its anti-tumor properties by targeting cancer cells. Thus four different cationic liposomes formulations were prepared by thin-film hydration method in optimal conditions: DOTAP (formulation A); DPPC/DOTAP (4:1 molar ratio, formulation B), DPPC/DMPC/DOTAP (4:1:1 molar ratio, formulation C) and DPPC/DMPC/DOTAP/DSPE-mPEG2000 (4:1:1:0.1 molar ratio, formulation D). They were studied to identify lipid-compositions offering effective GEM-entrapment and successful coating of FU on the liposome surface. Additional qualitative characteristics, such as particle size, polydispersity index, zeta potential, stability and in vitro drug release were then evaluated. Formulation C gave the best GEM-entrapment efficiency (EE) but formed aggregates when coated with FU, giving non-homogenous large size particles then not suitable for effective delivery. It was the same situation with formulation A and B. Only the formulation D showed a good GEM-EE (> 80%) and affinity by successful coating FU from three different algae species. The PEGylated formulation D coated of FU, with regard to storage stability and drug release studies, revealed to be a promising approach on design of optimal drug delivery system.
Collapse
Affiliation(s)
- Epiphane K Silli
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | | | - Xintao Zhou
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Mengfei Li
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Jiali Tang
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Ruizhe Guo
- School of Chinese Medicine Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Chunlu Tan
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ying Wang
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
40
|
Ferrara B, Dugnani E, Citro A, Schiavo Lena M, Marra P, Camisa PR, Policardi M, Canu T, Esposito A, Doglioni C, Piemonti L. ASO Author Reflections: Bridging the Gap in PDAC Research: The Intraportal Model as a Platform for Studying Preclinical Liver Metastasis. Ann Surg Oncol 2024; 31:7066-7067. [PMID: 38904860 DOI: 10.1245/s10434-024-15655-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/22/2024]
Affiliation(s)
- Benedetta Ferrara
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Erica Dugnani
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antonio Citro
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Schiavo Lena
- Pathology Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Marra
- Department of Radiology, Papa Giovanni XXIII Hospital, Bergamo, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Riccardo Camisa
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Martina Policardi
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Tamara Canu
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antonio Esposito
- Vita-Salute San Raffaele University, Milan, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Claudio Doglioni
- Pathology Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
41
|
Giuliante F, Panettieri E, Campisi A, Coppola A, Vellone M, De Rose AM, Ardito F. Treatment of oligo-metastatic pancreatic ductal adenocarcinoma to the liver: is there a role for surgery? A narrative review. Int J Surg 2024; 110:6163-6169. [PMID: 38818688 PMCID: PMC11486948 DOI: 10.1097/js9.0000000000001665] [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: 12/30/2023] [Accepted: 05/09/2024] [Indexed: 06/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a prognostically unfavorable malignancy that presents with distant metastases at the time of diagnosis in half of patients. Even if patients with metastatic PDAC have not been traditionally considered candidates for surgery, an increasing number of researchers have been investigating the efficacy of surgical treatment for patients with liver-only oligometastases from PDAC, showing promising results in extremely selected patients, mainly with metachronous metastases after perioperative chemotherapy. Nevertheless, a standardized definition of oligometastatic disease should be adopted and additional investigations focusing on the role of perioperative chemotherapy and tumor biology are warranted to reliably assess the role of resection for PDAC metastatic to the liver.
Collapse
Affiliation(s)
- Felice Giuliante
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| | - Elena Panettieri
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrea Campisi
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| | | | - Maria Vellone
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| | - Agostino M. De Rose
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| | - Francesco Ardito
- Hepatobiliary Surgery, Fondazione Policlinico Universitario Agostino Gemelli ‘IRCCS’, Università Cattolica del Sacro Cuore
| |
Collapse
|
42
|
Sousa SM, Branco H, Avan A, Palmeira A, Morelli L, Santos LL, Giovannetti E, Vasconcelos MH, Xavier CPR. Darifenacin: a promising chitinase 3-like 1 inhibitor to tackle drug resistance in pancreatic ductal adenocarcinoma. Cancer Chemother Pharmacol 2024; 94:585-597. [PMID: 39225813 PMCID: PMC11438711 DOI: 10.1007/s00280-024-04712-1] [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: 04/23/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is among the most aggressive malignancies. Our previous work revealed Chitinase 3-like 1 (CHI3L1) involvement in PDAC resistance to gemcitabine, identifying it as a promising therapeutic target. Here, we aimed to identify putative CHI3L1 inhibitors and to investigate their chemosensitizing potential in PDAC. METHODS Docking analysis for CHI3L1 identified promising CHI3L1 inhibitors, including darifenacin (muscarinic receptor antagonist). PDAC cell lines (BxPC-3, PANC-1) and primary PDAC cells were used to evaluate darifenacin's effects on cell growth (Sulforhodamine B, SRB), alone or in combination with gemcitabine or gemcitabine plus paclitaxel. Cytotoxicity against normal immortalized pancreatic ductal cells (HPNE) was assessed. Recombinant protein was used to confirm the impact of darifenacin on CHI3L1-induced PDAC cellular resistance to therapy (SRB assay). Darifenacin's effect on Akt activation was analysed by ELISA. The association between cholinergic receptor muscarinic 3 (CHRM3) expression and therapeutic response was evaluated by immunohistochemistry of paraffin-embedded tissues from surgical resections of a 68 patients' cohort. RESULTS In silico screening revealed the ability of darifenacin to target CHI3L1 with high efficiency. Darifenacin inhibited PDAC cell growth, with a GI50 of 26 and 13.6 µM in BxPC-3 and PANC-1 cells, respectively. These results were confirmed in primary PDAC-3 cells, while darifenacin showed no cytotoxicity against HPNE cells. Importantly, darifenacin sensitized PDAC cells to standard chemotherapies, reverted CHI3L1-induced PDAC cellular resistance to therapy, and decreased Akt phosphorylation. Additionally, high CHMR3 expression was associated with low therapeutic response to gemcitabine. CONCLUSION This work highlights the potential of darifenacin as a chemosensitizer for PDAC treatment.
Collapse
Affiliation(s)
- Sofia M Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- LQOF - Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Helena Branco
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, 91886-17871, Iran
- Medical Genetics Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, 91886-17871, Iran
| | - Andreia Palmeira
- LQOF - Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
- CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, Terminal de Cruzeiros do Porto de Leixões, Matosinhos, 4450-208, Portugal
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, 56100, Italy
| | - Lúcio L Santos
- Experimental Pathology and Therapeutics Research Group and Surgical Oncology Department, IPO-Instituto Português de Oncologia, Rua Dr. António Bernardino de Almeida 865, Porto, 4200-072, Portugal
- ICBAS-UP-School of Medicine and Biomedical Sciences, University of Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050- 313, Portugal
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, HV Amsterdam, 1081, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana per La Scienza, San Giuliano, 56017, Italy
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal.
- Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal.
- Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal.
| | - Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal.
- Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal.
- UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Gandra, 4585-116, Portugal.
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, Gandra, 4585-116, Portugal.
| |
Collapse
|
43
|
Li M, Freeman S, Franco-Barraza J, Cai KQ, Kim A, Jin S, Cukierman E, Ye K. A bioprinted sea-and-island multicellular model for dissecting human pancreatic tumor-stroma reciprocity and adaptive metabolism. Biomaterials 2024; 310:122631. [PMID: 38815457 PMCID: PMC11186049 DOI: 10.1016/j.biomaterials.2024.122631] [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: 01/11/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents a formidable clinical challenge due to its intricate microenvironment characterized by desmoplasia and complex tumor-stroma interactions. Conventional models hinder studying cellular crosstalk for therapeutic development. To recapitulate key features of PDAC masses, this study creates a novel sea-and-island PDAC tumor construct (s&i PTC). The s&i PTC consists of 3D-printed islands of human PDAC cells positioned within an interstitial extracellular matrix (ECM) populated by human cancer-associated fibroblasts (CAFs). This design closely mimics the in vivo desmoplastic architecture and nutrient-poor conditions. The model enables studying dynamic tumor-stroma crosstalk and signaling reciprocity, revealing both known and yet-to-be-discovered multicellular metabolic adaptations. Using the model, we discovered the orchestrated dynamic alterations of CAFs under nutrient stress, resembling critical in vivo human tumor niches, such as the secretion of pro-tumoral inflammatory factors. Additionally, nutrient scarcity induces dynamic alterations in the ECM composition and exacerbates poor cancer cell differentiation-features well-established in PDAC progression. Proteomic analysis unveiled the enrichment of proteins associated with aggressive tumor behavior and ECM remodeling in response to poor nutritional conditions, mimicking the metabolic stresses experienced by avascular pancreatic tumor cores. Importantly, the model's relevance to patient outcomes is evident through an inverse correlation between biomarker expression patterns in the s&i PTCs and PDAC patient survival rates. Key findings include upregulated MMPs and key ECM proteins (such as collagen 11 and TGFβ) under nutrient-avid conditions, known to be regulated by CAFs, alongside the concomitant reduction in E-cadherin expression associated with a poorly differentiated PDAC state under nutrient deprivation. Furthermore, elevated levels of hyaluronic acid (HA) and integrins in response to nutrient deprivation underscore the model's fidelity to the PDAC microenvironment. We also observed increased IL-6 and reduced α-SMA expression under poor nutritional conditions, suggesting a transition of CAFs from myofibroblastic to inflammatory phenotypes under a nutrient stress akin to in vivo niches. In conclusion, the s&i PTC represents a significant advancement in engineering clinically relevant 3D models of PDAC masses. It offers a promising platform for elucidating tumor-stroma interactions and guiding future therapeutic strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Ming Li
- Department of Biomedical Engineering, Center of Biomanufacturing for Regenerative Medicine, Binghamton University, SUNY, Binghamton, NY, USA
| | - Sebastian Freeman
- Department of Biomedical Engineering, Center of Biomanufacturing for Regenerative Medicine, Binghamton University, SUNY, Binghamton, NY, USA
| | - Janusz Franco-Barraza
- Cancer Signaling and Microenvironment Program, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz Temple School of Medicine, Philadelphia, PA, USA
| | - Kathy Q Cai
- Cancer Signaling and Microenvironment Program, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz Temple School of Medicine, Philadelphia, PA, USA
| | - Amy Kim
- Department of Biomedical Engineering, Center of Biomanufacturing for Regenerative Medicine, Binghamton University, SUNY, Binghamton, NY, USA
| | - Sha Jin
- Department of Biomedical Engineering, Center of Biomanufacturing for Regenerative Medicine, Binghamton University, SUNY, Binghamton, NY, USA
| | - Edna Cukierman
- Cancer Signaling and Microenvironment Program, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz Temple School of Medicine, Philadelphia, PA, USA.
| | - Kaiming Ye
- Department of Biomedical Engineering, Center of Biomanufacturing for Regenerative Medicine, Binghamton University, SUNY, Binghamton, NY, USA.
| |
Collapse
|
44
|
Carlomagno S, Setti C, Ortolani F, Sivori S. Pancreatic ductal adenocarcinoma microenvironment: Soluble factors and cancer associated fibroblasts as modulators of NK cell functions. Immunol Lett 2024; 269:106898. [PMID: 39019404 DOI: 10.1016/j.imlet.2024.106898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is the most frequent pancreatic cancer and represents one of the most aggressive human neoplasms. Typically identified at advance stage disease, most PDAC tumors are unresectable and resistant to standard therapies. The immunosuppressive microenvironment in PDAC impedes tumor control but a greater understanding of the complex stromal interactions within the tumor microenvironment (TME) and the development of strategies capable of restoring antitumor effector immune responses could be crucial to fight this aggressive tumor and its spread. Natural Killer (NK) cells play a crucial role in cancer immunosurveillance and represent an attractive target for immunotherapies, both as cell therapy and as a pharmaceutical target. This review describes some crucial components of the PDAC TME (collagens, soluble factors and fibroblasts) that can influence the presence, phenotype and function of NK cells in PDAC patients tumor tissue. This focused overview highlights the therapeutic relevance of dissecting the complex stromal composition to define new strategies for NK cell-based immunotherapies to improve the treatment of PDAC.
Collapse
Affiliation(s)
- Simona Carlomagno
- Department of Medicine (DMED), University of Udine, Piazzale Kolbe 4, Udine 33100, Italy.
| | - Chiara Setti
- Department of Experimental Medicine (DIMES), University of Genoa, Via Leon Battista Alberti 2, Genoa 16132, Italy
| | - Fulvia Ortolani
- Department of Medicine (DMED), University of Udine, Piazzale Kolbe 4, Udine 33100, Italy
| | - Simona Sivori
- Department of Experimental Medicine (DIMES), University of Genoa, Via Leon Battista Alberti 2, Genoa 16132, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
45
|
Ma Y, Yang Y, Zhang H, Mugaanyi J, Hu Y, Wu S, Lu C, Mao S, Wang K. Sarcomatoid carcinoma of the pancreas (Review). Oncol Lett 2024; 28:477. [PMID: 39161336 PMCID: PMC11332573 DOI: 10.3892/ol.2024.14610] [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: 10/31/2023] [Accepted: 06/20/2024] [Indexed: 08/21/2024] Open
Abstract
Sarcomatoid carcinoma of the pancreas (SCP) is a rare and aggressive subtype of undifferentiated pancreatic ductal adenocarcinoma, with a generally poor prognosis and only sporadic cases reported worldwide. Histologically, the most notable feature of SCP is the presence of abundant of mesenchymatoid spindle tumor cells in the tumor, which lack glandular differentiation. Immunohistochemically, SCP is characterized by the expression of both mesenchymal and epithelial markers. With only a few reported cases, there is limited knowledge about its molecular and clinicopathological characteristics. Therefore, the present study performed a literature search to identify all relevant published studies. The present review provides an overview of the histogenesis, diagnosis, genetic features, prognosis and treatment of SCP, specifically focusing on the molecular alterations. Furthermore, a single-center experience is reported, which adds to the limited evidence available in the literature.
Collapse
Affiliation(s)
- Yijie Ma
- Department of Hepatobiliary Pancreatic Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang 315048, P.R. China
| | - Yiwen Yang
- Department of Hepatobiliary Pancreatic Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang 315048, P.R. China
| | - Huizhi Zhang
- Department of Hepatobiliary Pancreatic Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang 315048, P.R. China
| | - Joseph Mugaanyi
- Department of Hepatobiliary Pancreatic Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang 315048, P.R. China
| | - Yangke Hu
- Department of Hepatobiliary Pancreatic Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang 315048, P.R. China
| | - Shengdong Wu
- Department of Hepatobiliary Pancreatic Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang 315048, P.R. China
| | - Caide Lu
- Department of Hepatobiliary Pancreatic Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang 315048, P.R. China
| | - Shuqi Mao
- Department of Hepatobiliary Pancreatic Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang 315048, P.R. China
| | - Ke Wang
- Department of Hepatobiliary Pancreatic Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang 315048, P.R. China
| |
Collapse
|
46
|
Kim HG, Cho Y, Lee JS, Oh ET, Park HJ. Identification of miR-6794-3p as a suppressor in pancreatic cancer metastasis. Int J Biol Sci 2024; 20:5272-5292. [PMID: 39430246 PMCID: PMC11488588 DOI: 10.7150/ijbs.98490] [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: 05/15/2024] [Accepted: 09/14/2024] [Indexed: 10/22/2024] Open
Abstract
Metastasis is a major cause of treatment failure in patients with pancreatic cancer, highlighting the urgent need for effective therapeutic strategies. Here, we focused on identifying novel miRNAs with key roles in metastasis of pancreatic cancer. Microarray analysis of miRNA expression in metastatic and non-metastatic pancreatic cancer samples revealed significantly lower expression of miR-6794-3p in the metastatic tumor group. Gain- and loss-of-function approaches using the pancreatic cancer cell lines MIA-PaCa-2 and HPAF-II expressing low and high levels of miR-6794-3p, respectively, indicated a role of miR-6794-3p in suppression of cell invasion, migration, and EMT signaling. Importantly, our results showed that miR-6794-3p exerts its effects by inhibiting expression of the chromatin remodeling factor, RBBP4. The resulting suppression of RBBP4 induced an increase in the levels of GRHL2 involved in regulating invasion, migration, and EMT signaling in metastatic pancreatic cancer cells. Consistent with these findings, low miR-6794-3p expression levels correlate with poor pancreatic cancer patient survival. Additional preclinical experiments on nude mice clearly demonstrated inhibitory effects of miR-6794-3p on pancreatic cancer cell metastasis. The collective results highlight the functional significance of miR-6794-3p as a suppressor of metastasis and support its predictive utility as a prognostic biomarker and therapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Ha Gyeong Kim
- Department of Microbiology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Yunmi Cho
- Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Jae-Seon Lee
- Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Republic of Korea
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Research Center for Controlling Intracellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Eun-Taex Oh
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Heon Joo Park
- Department of Microbiology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Republic of Korea
- Research Center for Controlling Intracellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
47
|
Munnings R, Gibbs P, Lee B. Evolution of Liquid Biopsies for Detecting Pancreatic Cancer. Cancers (Basel) 2024; 16:3335. [PMID: 39409954 PMCID: PMC11475855 DOI: 10.3390/cancers16193335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy characterised by late diagnosis and poor prognosis. Despite advancements, current diagnostic and prognostic strategies remain limited. Liquid biopsy techniques, including circulating tumour DNA (ctDNA), circulating tumour cells (CTCs), circulating tumour exosomes, and proteomics, offer potential solutions to improve PDAC diagnosis, prognostication, and management. A systematic search of Ovid MEDLINE identified studies published between 2019 and 2024, focusing on liquid biopsy biomarkers for PDAC. A total of 49 articles were included. ctDNA research shows some promise in diagnosing and prognosticating PDAC, especially through detecting mutant KRAS in minimal residual disease assays. CTC analyses had low sensitivity for early-stage PDAC and inconsistent prognostic results across subpopulations. Exosomal studies revealed diverse biomarkers with some diagnostic and prognostic potential. Proteomics, although relatively novel, has demonstrated superior accuracy in PDAC diagnosis, including early detection, and notable prognostic capacity. Proteomics combined with CA19-9 analysis has shown the most promising results to date. An update on multi-cancer early detection testing, given its significance for population screening, is also briefly discussed. Liquid biopsy techniques offer promising avenues for improving PDAC diagnosis, prognostication, and management. In particular, proteomics shows considerable potential, yet further research is needed to validate existing findings and comprehensively explore the proteome using an unbiased approach.
Collapse
Affiliation(s)
- Ryan Munnings
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
- Department of Medical Education, Melbourne Medical School, Parkville, VIC 3052, Australia
| | - Peter Gibbs
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
- Western Health, Footscray, VIC 3011, Australia
| | - Belinda Lee
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
- Peter MacCallum Cancer Centre, Parkville, VIC 3052, Australia
- Northern Health, Epping, VIC 3076, Australia
| |
Collapse
|
48
|
Khalid A, Pasha SA, Demyan L, Standring O, Newman E, King DA, DePeralta D, Gholami S, Weiss MJ, Melis M. Modified 5-Item Frailty Index (mFI-5) may predict postoperative outcomes after pancreatoduodenectomy for pancreatic Cancer. Langenbecks Arch Surg 2024; 409:286. [PMID: 39305322 DOI: 10.1007/s00423-024-03483-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 09/17/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Pancreatic Ductal Adenocarcinoma (PDAC) primarily affects older individuals with diminished physiological reserves. The Modified 5-Item Frailty Index (mFI-5) is a novel risk stratification tool proposed to predict postoperative morbidity and mortality. This study aimed to validate the mFI-5 for predicting surgical outcomes in patients undergoing pancreatoduodenectomy (PD) for PDAC. METHODS Our retrospective PDAC database included patients who underwent PD between 2014 and 2023. Patients were stratified by mFI-5 scores (0 best - 5 worst), which assess preoperative CHF, diabetes mellitus, history of COPD or pneumonia, functional health status, and hypertension requiring medication. Associations between mFI-5 scores and outcomes, including postoperative complications and mortality, were analyzed using logistic regression, Cox proportional hazards models, and Kaplan-Meier survival analysis. RESULTS Among 250 PDAC patients undergoing PD, 142 (56.8%) had mFI-5 scores ≤ 1, and 25 (10%) had scores ≥ 3. No patients had scores > 4. Higher mFI-5 scores correlated with older age (p < 0.001) and tobacco use (p = 0.036). Multivariate analysis identified age (RR 1.02, p = 0.038), ASA class (ASA III; RR 2.61, p < 0.001; ASA IV; RR 2.63, p = 0.026), and moderate alcohol consumption (RR 0.56, p = 0.038) as frailty predictors. An mFI-5 score > 2 independently associated with higher mortality (HR 2.08, p = 0.026). Median overall survival was significantly lower for patients with mFI-5 scores > 2 than for those with scores ≤ 2 (21.3 vs. 42.1 months, p = 0.022). CONCLUSIONS The mFI-5 is a valuable tool for predicting postoperative morbidity and mortality in PDAC patients undergoing PD. Integrating frailty assessment into preoperative evaluations can enhance patient selection and surgical outcomes. Future research should focus on incorporating frailty assessments into surgical planning and patient management to improve outcomes in this vulnerable population.
Collapse
Affiliation(s)
- Abdullah Khalid
- North Shore/Long Island Jewish General Surgery, Northwell Health, 300 Community Dr. Manhasset, Manhasset, NY, USA.
| | - Shamsher A Pasha
- Department of Surgery, UT Health San Antonio, San Antonio, TX, USA
| | - Lyudmyla Demyan
- North Shore/Long Island Jewish General Surgery, Northwell Health, 300 Community Dr. Manhasset, Manhasset, NY, USA
| | - Oliver Standring
- North Shore/Long Island Jewish General Surgery, Northwell Health, 300 Community Dr. Manhasset, Manhasset, NY, USA
| | - Elliot Newman
- Northwell Health Lenox Hill Hospital, 100 E 77th St, New York, NY, USA
| | - Daniel A King
- Northwell Health Cancer Institute, 1111 Marcus Ave, New Hyde Park, NY, USA
| | - Danielle DePeralta
- Northwell Health Cancer Institute, 1111 Marcus Ave, New Hyde Park, NY, USA
| | - Sepideh Gholami
- Northwell Health Cancer Institute, 1111 Marcus Ave, New Hyde Park, NY, USA
| | - Matthew J Weiss
- Northwell Health Cancer Institute, 1111 Marcus Ave, New Hyde Park, NY, USA
| | | |
Collapse
|
49
|
Szydlak R. Synergistic Inhibition of Pancreatic Cancer Cell Growth and Migration by Gemcitabine and Withaferin A. Biomolecules 2024; 14:1178. [PMID: 39334944 PMCID: PMC11430445 DOI: 10.3390/biom14091178] [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: 08/18/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Pancreatic cancer remains one of the most lethal malignancies due to its aggressive nature and resistance to conventional therapies. This study investigates the anti-proliferative, pro-apoptotic, and anti-migratory effects of Gemcitabine (GC) and Withaferin A (WFA) on pancreatic cancer cell lines PANC-1 and Hs766t. The MTS assay revealed that both compounds effectively inhibit cell proliferation, with WFA showing a stronger effect in Hs766t cells. Flow cytometry analysis demonstrated that GC and WFA, particularly in combination, significantly induce apoptosis in both cell lines. Migration assays confirmed the potent inhibition of cell migration by both compounds, with the combination treatment being the most effective. Furthermore, actin cytoskeleton analysis indicated substantial changes in cell morphology and stiffness, suggesting that GC and WFA disrupt the structural integrity of cancer cells. Additionally, the study highlights a ROS-mediated mechanism underlying the effects of GC and WFA, as evidenced by increased ROS levels following treatment, which were attenuated by N-acetylcysteine. Importantly, NF-κB activity was significantly modulated, with WFA reducing NF-κB activation induced by GC, potentially contributing to the synergistic pro-apoptotic effect of the combination. These findings suggest that the combination of GC and WFA may offer a synergistic therapeutic approach for treating pancreatic cancer by targeting multiple aspects of tumor cell behavior.
Collapse
Affiliation(s)
- Renata Szydlak
- Department of Bioinformatics and Telemedicine, Faculty of Medicine, Jagiellonian University Medical College, Medyczna 7, 30-688 Krakow, Poland
| |
Collapse
|
50
|
Hou Z, Lu F, Lin J, Wu Y, Chen L, Fang H, Chen L, Zhang S, Huang H, Pan Y. Loss of Annexin A1 in macrophages restrains efferocytosis and remodels immune microenvironment in pancreatic cancer by activating the cGAS/STING pathway. J Immunother Cancer 2024; 12:e009318. [PMID: 39237260 PMCID: PMC11381726 DOI: 10.1136/jitc-2024-009318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2024] [Indexed: 09/07/2024] Open
Abstract
OBJECTIVE Pancreatic cancer is an incurable malignant disease with extremely poor prognosis and a complex tumor microenvironment. We sought to characterize the role of Annexin A1 (ANXA1) in pancreatic cancer, including its ability to promote efferocytosis and antitumor immune responses. METHODS The tumor expression of ANXA1 and cleaved Caspase-3 (c-Casp3) and numbers of tumor-infiltrating CD68+ macrophages in 151 cases of pancreatic cancer were examined by immunohistochemistry and immunofluorescence. The role of ANXA1 in pancreatic cancer was investigated using myeloid-specific ANXA1-knockout mice. The changes in tumor-infiltrating immune cell populations induced by ANXA1 deficiency in macrophages were assessed by single-cell RNA sequencing and flow cytometry. RESULTS ANXA1 expression in pancreatic cancer patient samples correlated with the number of CD68+ macrophages. The percentage of ANXA1+ tumor-infiltrating macrophages negatively correlated with c-Casp3 expression and was significantly associated with worse survival. In mice, myeloid-specific ANXA1 deficiency inhibited tumor growth and was accompanied by the accumulation of apoptotic cells in pancreatic tumor tissue caused by inhibition of macrophage efferocytosis, which was dependent on cGAS-STING pathway-induced type I interferon signaling. ANXA1 deficiency significantly remodeled the intratumoral lymphocyte and macrophage compartments in tumor-bearing mice by increasing the number of effector T cells and pro-inflammatory macrophages. Furthermore, combination therapy of ANXA1 knockdown with gemcitabine and anti-programmed cell death protein-1 antibody resulted in synergistic inhibition of pancreatic tumor growth. CONCLUSION This research uncovers a novel role of macrophage ANXA1 in pancreatic cancer. ANXA1-mediated regulation of efferocytosis by tumor-associated macrophages promotes antitumor immune response via STING signaling, suggesting potential treatment strategies for pancreatic cancer.
Collapse
Affiliation(s)
- Zelin Hou
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Fengchun Lu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jiajing Lin
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yuwei Wu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Linjin Chen
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Haizong Fang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Linlin Chen
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shihan Zhang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Heguang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yu Pan
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- The Cancer Center, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|