51
|
Dunn M, Dymock L, Hoskins C. Solid lipid nanoparticles in pancreatic cancer treatment. BJC REPORTS 2025; 3:21. [PMID: 40217114 PMCID: PMC11992092 DOI: 10.1038/s44276-025-00130-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/13/2025] [Accepted: 03/02/2025] [Indexed: 04/14/2025]
Abstract
Pancreatic cancer comes with one of the poorest prognoses of all cancers and as such it is crucial that new therapies are developed to improve on the current statistics. Currently, chemotherapy is the cornerstone of pancreatic cancer treatment with several drugs, and combinations of drugs being utilised for their anti-cancer effect. However, pancreatic cancer has a dense stroma around the tumour and intratumoral bacteria which result in drugs having difficulty penetrating the tumour or being metabolised by bacteria rendering them inactive. The utilisation of nanotechnology in chemotherapy for pancreatic cancer has been a huge area of focus for researchers worldwide with most of the focus being on lipid-based, inorganic and polymer-based nanoparticles. Solid lipid nanoparticles which have been studied since being first published in the 1990s, have been poorly researched for pancreatic cancer applications. Being composed of physiological lipids, solid lipid nanoparticles offer a greatly reduced risk of acute or chronic toxicities arising compared to inorganic or polymeric nanoparticles. They also possess the ability to improve on circulation time, permeability, and bioavailability of many first-line chemotherapeutics.
Collapse
Affiliation(s)
- Mia Dunn
- Department of Pure and Applied Chemistry, University of Strathclyde, Technology Innovation Centre, 99 George Street, Glasgow, G1 1RD, UK
| | - Lewis Dymock
- Department of Pure and Applied Chemistry, University of Strathclyde, Technology Innovation Centre, 99 George Street, Glasgow, G1 1RD, UK
| | - Clare Hoskins
- Department of Pure and Applied Chemistry, University of Strathclyde, Technology Innovation Centre, 99 George Street, Glasgow, G1 1RD, UK.
| |
Collapse
|
52
|
Jin Y, Tao H, Liu Y, Liu S, Tang X. LINC00704 boosts the immunologic escape of colorectal cancer cells by upregulating TLR4 by binding with miR- 203a- 3p. Eur J Med Res 2025; 30:263. [PMID: 40211393 PMCID: PMC11983970 DOI: 10.1186/s40001-025-02514-6] [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: 12/23/2024] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a common malignant tumor and is the second most common cause of cancer-related deaths worldwide. Immune escape suppresses anti-tumor immunity and facilitates tumor cells to proliferate. MiR- 203a- 3p regulates cancer progression and LINC00704 may bind with miR- 203a- 3p to inhibit its effects. METHODS In this study, the levels of miR- 203a- 3p and LINC00704 were tested in tumor tissue and non-cancer tissues in vivo. In further in vitro experiments, transfection, cell vitality, apoptosis, and proliferation ability were detected. The expression level of TLR4 was also examined. Finally, a luciferase assay was conducted to detect whether LINC00704 could bind with miR- 203a- 3p. RESULTS A rise in LINC00704 mRNA was observed in CRC tissues while miR- 203a- 3p was reduced. LINC00704 boosts the proliferation of cells and inhibits cell apoptosis. LINC00704 regulates Toll- 1ike receptor- 4 (TLR4) expression through miR- 203a- 3p, thereby modulating cell viability. CRC cell immune escape was facilitated by LINC00704 via miR- 203a- 3p. CONCLUSION LINC00704 promotes CRC cell immunologic escape by upgrading TLR4 by binding with miR- 203a- 3p.
Collapse
Affiliation(s)
- Yalei Jin
- Dept. of General Practice, Zhongnanhongnan Hospital of Wuhanuhan Universityniversity, No169 Donghu Road, Wuchang District, Wuhan, Hubei, China
| | - Hai Tao
- Dept. of Orthopedics, Renmin Hospital of Wuhan University, No 99 Zhangzhidong Street, Wuchang District, Wuhan, Hubei, China
| | - Yuwei Liu
- Dept. of General Practice, Zhongnanhongnan Hospital of Wuhanuhan Universityniversity, No169 Donghu Road, Wuchang District, Wuhan, Hubei, China
| | - Sha Liu
- Dept. of General Practice, Zhongnanhongnan Hospital of Wuhanuhan Universityniversity, No169 Donghu Road, Wuchang District, Wuhan, Hubei, China
| | - Xiaoyan Tang
- Dept. of General Practice, Zhongnanhongnan Hospital of Wuhanuhan Universityniversity, No169 Donghu Road, Wuchang District, Wuhan, Hubei, China.
| |
Collapse
|
53
|
Choi J, Gordon A, Eresen A, Zhang Z, Borhani A, Bagci U, Lewandowski R, Kim DH. Current applications of radiomics in the assessment of tumor microenvironment of hepatocellular carcinoma. Abdom Radiol (NY) 2025:10.1007/s00261-025-04916-w. [PMID: 40208284 DOI: 10.1007/s00261-025-04916-w] [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: 11/30/2024] [Revised: 02/10/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025]
Abstract
The tumor microenvironment (TME) of hepatocellular carcinoma (HCC) has garnered significant attention, especially with the rise of immunotherapy as a treatment strategy. Radiomics, an innovative technique, offers valuable insights into the intricate structure of the TME. This review highlights recent advancements in radiomics for analyzing the HCC TME, identifies key areas that warrant further research, and explores comprehensive multi-omics approaches that extend the potential of radiomics to new frontiers.
Collapse
Affiliation(s)
- Junghwa Choi
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Andrew Gordon
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Aydin Eresen
- Department of Radiological Sciences, University of California, Irvine, Irvine, USA
| | - Zhuoli Zhang
- Department of Radiological Sciences, University of California, Irvine, Irvine, USA
| | - Amir Borhani
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Ulas Bagci
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Robert Lewandowski
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, 60611, USA.
| |
Collapse
|
54
|
Elgedawy GA, Elabd NS, Elbrolosy AM, El-Morshedy SM, El-Gamal A, Abozeid M, Abdelkreem M, Eleowa SS, Helal ML. Circulating miR-485-5p as a potential diagnostic and prognostic biomarker for HCV-related hepatocellular carcinoma. Clin Exp Med 2025; 25:110. [PMID: 40208438 PMCID: PMC11985578 DOI: 10.1007/s10238-025-01625-y] [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: 12/13/2024] [Accepted: 03/05/2025] [Indexed: 04/11/2025]
Abstract
Hepatitis C virus (HCV) is the predominant viral cause of hepatocellular carcinoma (HCC). Early detection and use of reliable biological markers can improve survival for HCC patients. MiR-485-5p was identified as a tumor-suppressing microribonucleic acid (miRNA) in some human cancers and was recently found to be downregulated in HCC tissues, signifying its utility as a promising biomarker. We aimed to investigate the potential role of circulating miR-485-5p as a novel diagnostic and prognostic biomarker for HCV-related HCC. This case-control study included 50 patients with HCC associated with HCV, 50 patients with HCV-related liver cirrhosis, and 50 healthy controls. History gathering, physical examination, laboratory, and imaging assessments were performed. A quantitative real-time polymerase chain reaction was used to measure miR-485-5p levels. Serum miR-485-5p values demonstrated a stepwise decline pattern from the control group to cirrhotic patients, with the HCC group exhibiting the lowest levels (p < 0.001). HCC patients with early BCLC stages had significantly lower miR-485-5p levels than those with late stages (p = 0.004). The miR-485-5p displayed a better performance in predicting HCV-related HCC with a greater area under the ROC curve (AUC) than alpha-fetoprotein (AFP) (AUC and sensitivity 0.921 and 92.0 versus 0.704 and 64.0, respectively) (p < 0.001). Also, its performance in predicting HCC prognosis surpassed that of AFP (AUC and sensitivity 0.872 and 85.19 versus 0.695 and 62.96, respectively) (p < 0.001). Circulating miR-485-5p is a promising, accurate, and noninvasive biomarker for the early detection and prediction of prognosis in patients with HCV-linked HCC.
Collapse
Affiliation(s)
- Gamalat A Elgedawy
- Clinical Biochemistry and Molecular Diagnostics Department, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Naglaa S Elabd
- Tropical Medicine Depatment, Faculty of Medicine, Menoufia University, Shebin El-Kom, Menoufia, 32511, Egypt.
| | - Asmaa M Elbrolosy
- Medical Microbiology and Immunology Department, Faculty of Medicine, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Suzan M El-Morshedy
- Clinical Pathology Department, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Ayman El-Gamal
- Tropical Medicine Depatment, Faculty of Medicine, Menoufia University, Shebin El-Kom, Menoufia, 32511, Egypt
| | - Mai Abozeid
- Hepatology and Gastroenterology Department, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, 32511, Egypt.
| | - Mervat Abdelkreem
- Hepatology and Gastroenterology Department, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, 32511, Egypt
| | - Sama S Eleowa
- BMS-University of Science and Technology at Zewail City, Giza, Egypt
| | - Marwa L Helal
- Clinical Biochemistry and Molecular Diagnostics Department, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| |
Collapse
|
55
|
Yu F, Zhang G, Sun J, Zhao Y, Qi Y, Han X, Ai C, Sun W, Duan J, Yu D. Nanotension Relief Agent Enhances Tissue Penetration by Reducing Solid Stress in Pancreatic Ductal Adenocarcinoma via Rho/ROCK Pathway Inhibition. Biomater Res 2025; 29:0173. [PMID: 40207257 PMCID: PMC11979343 DOI: 10.34133/bmr.0173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
The formidable contractile tension exerted by cancer-associated fibroblasts (CAFs) in pancreatic ductal adenocarcinoma (PDAC) tissue is crucial for maintaining high tissue solid stress (TSS), which impedes the delivery and penetration of chemotherapeutic drugs. To address this obstacle, we constructed a pH-responsive nanotension relief agent (FS@MMS), in which fasudil (FS) was ingeniously conjugated to mesoporous silica encapsulated with magnetic iron oxide (MMS). The nanotension relief agent was demonstrated to inhibit the synthesis of phosphorylated myosin light chain by blocking the Rho/Rho-associated serine/threonine kinase (ROCK) pathway, triggering the swift transformation of high-tension CAFs into low-tension CAFs in PDAC tissue, which relieves TSS and enhances drug penetration in Panc02/NIH-3T3 multicellular tumor spheroids. When the nanotension relief agent was further loaded with the chemotherapeutic drug gemcitabine (GEM), as FS@MMS-GEM, the enhanced permeation of GEM progressively killed tumor cells and amplified their TSS-relief properties, thereby maximizing the anticancer efficacy of chemotherapeutic agents in Panc02/NIH-3T3 coplanted model mice. The magnetic resonance imaging results revealed that the synergistic effect substantially improved drug delivery and penetration efficiency. The developed approach holds great potential for improving chemotherapy efficacy in PDAC and provides a novel therapeutic approach for the treatment of related stroma-rich tumors.
Collapse
Affiliation(s)
- Feiran Yu
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Gaorui Zhang
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jintang Sun
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yuxuan Zhao
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yafei Qi
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Xiaoyu Han
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Chen Ai
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Weikai Sun
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
- Research Center for Basic Medical Sciences,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jiazhi Duan
- Institute for Advanced Interdisciplinary Research,
University of Jinan, Jinan 250022, China
| | - Dexin Yu
- Department of Radiology,
Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
- Translational Medicine Research Center in Nano Molecular and Functional Imaging of Shandong University, Jinan 250012, China
| |
Collapse
|
56
|
Chang HH, Luo HL, Su YL, Fang FM, Wang CJ, Huang CC. Long-term outcome of radiation therapy for locoregional recurrence of upper tract urothelial carcinoma after nephroureterectomy. BMC Urol 2025; 25:80. [PMID: 40205429 PMCID: PMC11980227 DOI: 10.1186/s12894-025-01766-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND The objective was to evaluate the survival outcomes associated with radiation therapy (RT) in cases of postoperative locoregional recurrence of upper tract urothelial carcinoma (UTUC) following radical nephroureterectomy (RNU). METHODS A total of 30 patients undergoing RT for local or regional recurrence of UTUC after RNU from 2002 to 2017 were enrolled. The median follow-up time for patients who survived was 5.9 years. The application of RT comprised two intents: (1) salvage RT for patients who did not respond to chemotherapy or who did not receive chemotherapy, and (2) consolidation RT for patients who showed complete or partial response to chemotherapy. Overall survival (OS) since recurrence, locoregional progression-free survival (LRPFS) after RT, and distant metastasis-free survival (DMFS) after RT were evaluated. Univariate Cox regression analysis was applied. RESULTS The median OS since recurrence was 40.0 months, and the 5-year OS, LRPFS, and DMFS rates were 46.6%, 45.0%, and 36.7%, respectively. For 14 patients who underwent salvage RT, the 5-year OS, LRPFS, and DMFS rates were 28.6%, 21.4%, and 21.4%, respectively; for 16 patients undergoing consolidation RT, those rates were 63.5%, 65.8%, and 50.0%, respectively. In univariate analysis, consolidation RT and primary site in the ureter were significant prognostic factors for better OS and LRPFS; an age ≤ 60.0 years was also a significant factor for OS. There was no significant factor for DMFS. CONCLUSIONS The patients undergoing RT for postoperative locoregional recurrence of UTUC had promising survival outcomes. Future prospective randomized trials to verify the findings are needed.
Collapse
Affiliation(s)
- Ho-Hsiang Chang
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Show Chwan Memorial Hospital, Kaohsiung, Taiwan
| | - Hao-Lun Luo
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Li Su
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Fu-Min Fang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chong-Jong Wang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Radiation Oncology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Chun-Chieh Huang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
- Department of Radiation Oncology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan.
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
57
|
Wang F, Zhou H, Tian Y, Wang X, Huang Y, Tu Y, Li L, Zhen H. ELK4 induced upregulation of HOMER3 promotes the proliferation and metastasis in glioma via Wnt/β-catenin/EMT signaling pathway. Biol Direct 2025; 20:48. [PMID: 40205485 PMCID: PMC11980352 DOI: 10.1186/s13062-025-00643-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
Glioma is an aggressive brain tumor characterized by its high invasiveness, which complicates prognosis and contributes to patient resistance against various treatment options. The HOMER family, consisting of HOMER1, HOMER2, and HOMER3, has been implicated in various cancers, yet their specific roles in glioma remain inadequately understood. This study conducted a comprehensive pan-cancer analysis to evaluate the expression profiles of HOMER family members across different tumor types, utilizing data from public databases such as TCGA and GTEx. Our findings indicate significant dysregulation of HOMER1, HOMER2, and HOMER3 in multiple cancers, with HOMER3 emerging as a potential prognostic biomarker, particularly for lower-grade glioma. Elevated expression levels of HOMER3 were associated with shorter overall survival and disease-specific survival in LGG patients, supported by Cox regression analysis that confirmed HOMER3 as an independent prognostic factor. Furthermore, HOMER3 expression correlated positively with advanced clinical stages and key tumor markers. To elucidate the mechanisms behind HOMER3 dysregulation, we identified ELK4 as a transcription factor that binds to the HOMER3 promoter, promoting its expression in glioma cells. Functional assays demonstrated that silencing HOMER3 significantly reduced glioma cell proliferation and metastatic potential in vitro and in vivo, highlighting its oncogenic role. Additionally, HOMER3 was found to influence the Wnt/β-catenin/EMT signaling pathway, with knockdown resulting in altered expression of critical EMT markers. Collectively, our results indicated that HOMER3 plays a crucial role in glioma progression and metastasis, underscoring its potential as a therapeutic target and prognostic biomarker in glioma management.
Collapse
Affiliation(s)
- Furong Wang
- Department of Pathology, Huizhou Central People's Hospital, Huizhou, Guangdong, China
- Department of Pathology, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China
| | - Hui Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Yu Tian
- Science Research Center, Huizhou Central People's Hospital, No. 41 E Ling North Road, Huizhou, Guangdong, China
- Science Research Center, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China
| | - Xiaoling Wang
- Science Research Center, Huizhou Central People's Hospital, No. 41 E Ling North Road, Huizhou, Guangdong, China
- Science Research Center, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China
| | - Youcai Huang
- Science Research Center, Huizhou Central People's Hospital, No. 41 E Ling North Road, Huizhou, Guangdong, China
- Science Research Center, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China
| | - Yanyang Tu
- Science Research Center, Huizhou Central People's Hospital, No. 41 E Ling North Road, Huizhou, Guangdong, China.
- Science Research Center, Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, China.
- Huizhou Central People's Hospital Academy of Medical Sciences, Huizhou, China.
| | - Liwen Li
- Department of Bioscience, College of Life Sciences, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China.
| | - Haining Zhen
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, No. 127 Changle West Road, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
58
|
Tang J, Li X, Tang N, Lin X, Du Y, Zhang S, Li Q, Zhang Y, Zhang Y, Hang H, Qiu T, Qiu Y, Cheng H, Dai Z, Hong H, Wei W, He J, Yan C. CD44 identified as a diagnostic biomarker for highly malignant CA19-9 negative pancreatic cancer. Cancer Lett 2025; 622:217713. [PMID: 40216152 DOI: 10.1016/j.canlet.2025.217713] [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: 02/26/2025] [Revised: 03/25/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with limited diagnostic biomarkers. Carbohydrate antigen 19-9 (CA19-9) is a widely used clinical biomarker and is generally considered to correlate with PDAC malignancy. However, the relationship between CA19-9 expression levels and tumor aggressiveness remains underexplored. In this study, we report a biphasic relationship between CA19-9 expression levels and PDAC malignancy, where both negative (<5 U/mL) and high (>37 U/mL) CA19-9 levels are associated with increased tumor aggressiveness. We defined CA19-9 negative PDAC as tumors that lack CA19-9 expression intracellulary, on the cell membrane, and in secreted form. In PDAC cell lines and patient-derived organoids, CA19-9 negativity, confirmed by immunofluorescence, flow cytometry and ELISA, correlated with more aggressive behaviors. In PDAC patients, tumors from those with serum CA19-9 levels below 5 U/mL exhibited stronger metabolically activity, more immunosuppressive tumor microenvironment, and worse survival than CA19-9 positive tumors, with over 90 % showing absent CA19-9 expression by immunohistochemistry (IHC). Glycoproteomics profiling identified CD44 as a highly expressed biomarker in CA19-9 negative PDAC. Elevated CD44 expression effectively distinguished CA19-9 negative PDAC from both CA19-9 positive PDAC and CA19-9 negative benign pancreatic diseases, suggesting its potential as a diagnostic tool. Furthermore, we developed a radionuclide-labeled CD44 antibody 89Zr-1M2E3, which specifically recognized CA19-9 negative PDAC tumors in preclinical models using PET-CT imaging. These findings highlight CD44 as a promising biomarker and therapeutic target for diagnosing and treating CA19-9 negative PDAC.
Collapse
Affiliation(s)
- Jiatong Tang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiaoyang Li
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Neng Tang
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiawen Lin
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yixiang Du
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China
| | - Shuo Zhang
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Qi Li
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yifan Zhang
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yixuan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China
| | - Hexing Hang
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Tongtong Qiu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yudong Qiu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Institute of Pancreatology, Nanjing University, Nanjing, Jiangsu Province, China
| | - Hao Cheng
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Zhan Dai
- Nanjing Okay Biotechnology Co., Ltd, Nanjing, Jiangsu Provinve, China
| | - Hao Hong
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Wei Wei
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, Jiangsu Province, China.
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Institute of Pancreatology, Nanjing University, Nanjing, Jiangsu Province, China.
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu Province, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu Province, China; Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Institute of Pancreatology, Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
59
|
Tamba M, Osumi H, Ogura M, Fukuoka S, Okamura A, Kanamori J, Imamura Y, Yoshino K, Udagawa S, Wakatsuki T, Shinozaki E, Watanabe M, Yamaguchi K, Chin K, Ooki A. Real-world safety and efficacy of neoadjuvant docetaxel, cisplatin, and 5-fluorouracil therapy for locally advanced esophageal squamous cell carcinoma. BMC Cancer 2025; 25:636. [PMID: 40200210 PMCID: PMC11980314 DOI: 10.1186/s12885-025-14011-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/24/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy with docetaxel, cisplatin plus 5-FU (DCF) has become the new standard of care for locally advanced esophageal squamous cell carcinoma (ESCC). In a real-world setting, the efficacy, recurrence, and adverse events (AEs) remain unclear. METHODS This retrospective cohort study included 86 patients who received neoadjuvant DCF followed by esophagectomy for resectable ESCC. RESULTS Following neoadjuvant DCF treatment, 75 patients underwent R0 curative resection. At the median follow-up of 19.2 months, the median disease-free survival (DFS)/recurrence-free survival (RFS) was not yet reached, with estimated 3-year DFS/RFS rates of 65.2%, respectively. The incidence of primary tumor regression grading (TRG) grade 1a and pathological complete response (pCR) were 21.3% (16/75) and 14.7% (11/75), respectively. The estimated 1-year DFS/RFS rates were 93.8% for primary TRG grade 1a and 100% for pCR. Baseline elevated serum SCC-antigen levels were inversely associated with achieving primary TRG grade 1a or pCR. In 64 patients who did not achieve pCR, residual tumor cells in the lymph nodes (ypN; HR, 16.96; 95% CI, 2.11-136.12; P < 0.01) and Glasgow prognostic score (GPS; HR, 8.34; 95% CI, 1.73-40.31; P < 0.01) were independent predictors of shorter DFS/RFS. The most common grade ≥ 3 AEs were neutropenia (61.6%) and febrile neutropenia (26.7%), which were not associated with clinicopathological factors. The most common non-hematological AEs were appetite loss (9.3%), pulmonary embolism (8.1%), diarrhea (7.0%), and nausea (2.3%). Nine patients discontinued neoadjuvant DCF due to toxicities. CONCLUSIONS Neoadjuvant DCF was effective and well-tolerated in real-world ESCC patients. Primary TRG grade 1a or pCR showed a favorable DFS/RFS, while positive ypN and GPS were independent risk factors for worse DFS/RFS.
Collapse
Affiliation(s)
- Mikako Tamba
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Hiroki Osumi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Mariko Ogura
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Shota Fukuoka
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Akihiko Okamura
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Jun Kanamori
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yu Imamura
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Koichiro Yoshino
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Shohei Udagawa
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Takeru Wakatsuki
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Eiji Shinozaki
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Keisho Chin
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Akira Ooki
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
| |
Collapse
|
60
|
Beyaz H, Kavaz D, Rizaner N. Chitosan nanoparticle encapsulation of thymus capitatus essential oil: in vitro release, antioxidant, antibacterial activity and cytotoxicity in MDA-MB-231 cells. Pharm Dev Technol 2025:1-15. [PMID: 40163347 DOI: 10.1080/10837450.2025.2487255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/07/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Thymus capitatus (Th. Ca) is known to treat mouth ulcers and respiratory infections in Cyprus. However, antioxidant, antibacterial, and cytotoxic potential of Th. Ca. EO on MDA-MB-231 cells and its' encapsulation into nanoparticles has not been well studied. Therefore, we aimed to analyze the antioxidant, antibacterial, cytotoxic potential, loading efficiency, and in vitro release profile of both Th. Ca. EO and Chitosan Nanoparticle (Ch. Np) - Th. Ca. EO. GC-MS analysis revealed 53.97% carvacrol, 14.53% borneol, and 12.09% sabinene presence in EO. The loading efficiency of Th. Ca. EO into Ch. Np. was calculated as 35.27% and the in vitro release profile reached a maximum of 68% in pH 7 for two weeks. The Minimum Inhibitory Concentration (MIC) assay showed that E. coli had an MIC50 of 0.3215 mg/ml while B. subtilis had an MIC50 of 0.5304 mg/ml. The antioxidant activity of the EO was assessed by performing a DPPH assay with an IC50 = 440 μg/ml. Trypan Blue Assay revealed that 60 µg/ml Th. Ca. EO significantly reduced the cell viability of MDA-MB-231 cells by 10.7% at 48h and 20.06% at 72h. Overall, Ch. Np. - Th. Ca. EO has shown a promising formulation for the pharmaceutical industry.
Collapse
Affiliation(s)
- Huseyin Beyaz
- Bioengineering Department, Faculty of Engineering, Cyprus International University, Turkey
| | - Doga Kavaz
- Bioengineering Department, Faculty of Engineering, Cyprus International University, Turkey
- Biotechnology Research Centre, Cyprus International University, Nicosia, Turkey
| | - Nahit Rizaner
- Bioengineering Department, Faculty of Engineering, Cyprus International University, Turkey
- Biotechnology Research Centre, Cyprus International University, Nicosia, Turkey
| |
Collapse
|
61
|
Jin Y, Hu H, Tian Y, Xu H, Yu Q, Cheng L, Guo X, Wang Z, Huang X, Wang X, Wang G. The role of LncRNA-MANCR induced by HIF-1α drive the malignant progression of pancreatic cancer by targeting miRNA-494/SIRT1 signaling axis under hypoxic conditions. Cancer Gene Ther 2025:10.1038/s41417-025-00900-0. [PMID: 40195439 DOI: 10.1038/s41417-025-00900-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 03/08/2025] [Accepted: 03/25/2025] [Indexed: 04/09/2025]
Abstract
This study revealed the prospective biological role and fundamental mechanisms of hypoxia-induced lncRNA-MANCR (MANCR), which is notably upregulated in pancreatic cancer (PC). This work uncovered the potential biological function and underlying mechanisms of hypoxia-induced MANCR, which is significantly elevated in PC. Microarray assays confirmed MANCR expression in the tissues of patients with PC and patients with chronic pancreatitis (CP), which positively correlated with sirtuin-1 (SIRT1) mRNA levels. Chromatin immunoprecipitation and luciferase assays were employed to gauge binding within the hypoxia-inducible factor-1α (HIF-1α)/MANCR/miRNA-494/SIRT1 pathway. Additionally, the association between MANCR expression and the clinical outcomes of patients with PC was confirmed. MANCR is significantly upregulated in PC cells under hypoxic conditions, which is closely linked to poor prognosis in patients with PC. Depletion of MANCR repressed in vitro proliferation, migration, and invasion of PC cells and in vivo growth of PC xenograft tumours. We further demonstrated that MANCR is localised in the cytoplasm and competitively binds miR-494, which directly targets SIRT1. Mechanically, the overexpression of SIRT1 improved the stability of the HIF-1α protein through deacetylation, leading to enhanced HIF-1α assembly. Moreover, MANCR underwent transcriptional regulation by HIF-1α in a hypoxic setting. This modulation was ascribed to HIF-1α binding to hypoxia response elements present in the MANCR promoter sequence. Data revealed the potential possibility of feedback between MANCR and HIF-1α, which may be conducive to hypoxia-induced oncogenicity and PC tumorigenesis, thereby providing a suitable therapeutic target.
Collapse
Affiliation(s)
- Yan Jin
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
- Oncological and Laparoscopic Surgery Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao Hu
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Yitong Tian
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Han Xu
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Qiao Yu
- Ultrasound medicine department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Long Cheng
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Oncological and Laparoscopic Surgery Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyu Guo
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Oncological and Laparoscopic Surgery Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zongwei Wang
- School of Medicine, Stanford University, San Francisco, CA, USA
| | - Xiaoxu Huang
- Department of Gastrointestinal Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Xiaoming Wang
- Department of Hepatobiliary Surgery, The First Affiliated Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
- Oncological and Laparoscopic Surgery Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
62
|
Wang L, Li Q, Sun Y, Wang S, Fu X, Wang X, Zheng Y, Gao A, Sun Y, Li J. Tumor-derived immunoglobulin-like transcript 3 inhibition reshapes the immunosuppressive tumor microenvironment and potentiates programmed cell death ligand 1 blockade immunotherapy in lung adenocarcinoma. Transl Oncol 2025; 56:102381. [PMID: 40199156 PMCID: PMC12008602 DOI: 10.1016/j.tranon.2025.102381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 01/13/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025] Open
Abstract
The low response rate of current immune checkpoint inhibitors in cancer has necessitated the development of new immune targets. Survival and public databases analyses were performed to determine the clinical significance of immunoglobulin-like transcript 3 (ILT3). The impact of ILT3 and apolipoprotein E (APOE) on tumor-associated macrophage (TAM) recruitment and polarization were evaluated by transwell assay, flow cytometry (FCM), and real-time PCR, while their impact on T cell survival and cytotoxicity was detected by CFSE, apoptotic assay, FCM and ELISA. These pro-tumoural activity of (an ortholog of ILT3 in mouse) were verified in vivo models. Survival and public databases analyses revealed that high ILT3 expression was significantly associated with worse prognosis in lung adenocarcinoma (LUAD), but not in squamous cell carcinoma. The same association was observed with its ligand, APOE. In vitro assays demonstrated that tumor-derived ILT3/APOE promoted recruitment and M2-like polarization of TAMs in LUAD and directly inhibited T cell proliferation and cytotoxicity. In vivo knockdown of gp49b enhanced anti-tumor immunity and suppressed tumor progression by counteracting TAM- and dysfunctional T cell-induced tumor microenvironment immunosuppression. Furthermore, combined inhibition of gp49b and programmed cell death ligand 1 (PD-L1) showed the most drastic tumor regression in C57BL/6 mice models. Tumor-derived ILT3 overexpression suppresses anti-tumor immunity by recruiting M2-like TAMs and impairing T cell activities, while ILT3 inhibition counteracts this immunosuppression and enhances the efficacy of PD-L1 blockade in LUAD. Thus, ILT3 could be a promising novel immunotherapeutic target for combined immunotherapy.
Collapse
Affiliation(s)
- Leirong Wang
- Phase I Clinical Research Center, Shandong University Cancer Center, Jinan, Shandong, China; Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qing Li
- Department of Oncology, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, Shandong, China
| | - Yanxin Sun
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shuyun Wang
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xuebing Fu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiufen Wang
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yan Zheng
- Jinan Center Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Aiqin Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yuping Sun
- Phase I Clinical Research Center, Shandong University Cancer Center, Jinan, Shandong, China; Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Juan Li
- Phase I Clinical Research Center, Shandong University Cancer Center, Jinan, Shandong, China; Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
63
|
Zhou C, Wang J, Zhou L, Li H, Liu X, Wang S, Zhang X, Ye X, Ren H, Zeng K, Li X, Wang D, Ji J. The novel Piperine derivative YL-1-9 exhibits anti-breast Cancer effects by inducing apoptosis via the p53/p21 pathway. Bioorg Med Chem Lett 2025; 123:130231. [PMID: 40204112 DOI: 10.1016/j.bmcl.2025.130231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/25/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
The tumor suppressor protein p53 plays a crucial role in the pathogenesis of breast cancer; however, its function is often compromised due to MDM2 overexpression or mutations in the p53 gene, which occurs in approximately 30-35 % of breast cancer cases. Piperine, a natural bioactive compound, has shown potential in inhibiting breast cancer cell growth by upregulating p53 expression. However, its clinical application is hindered by poor bioavailability, potential toxicity, and the risk of undesirable drug interactions. In the present study, a novel derivative of Piperine, YL-1-9, was synthesized and evaluated for its anticancer activity against breast cancer. YL-1-9, a bicyclic amide derivative of Piperine, was evaluated for antitumor effects both in vitro and in vivo using MTT assays and the chick embryo chorioallantoic membrane (CAM) model. Further investigations into its effects on breast cancer cell clonogenicity, adhesion, invasion, and migration were conducted through colony formation assays, EdU assays, cell adhesion and invasion studies, and wound healing experiments. Western blot analysis was performed to elucidate the effects of YL-1-9 on the cell cycle and apoptosis, which were further validated using YO-PRO-1 and propidium iodide dual staining. YL-1-9 significantly inhibited breast cancer cell proliferation, adhesion, invasion, and migration, while inducing cell cycle arrest and promoting apoptosis. Mechanistically, YL-1-9 downregulated critical proteins in the CDK4/6-cyclin D-Rb-E2F pathway and the Caspase 3/Bax/Bcl-2 apoptosis signaling pathway. These findings position YL-1-9 as a promising candidate for breast cancer therapy; however, further clinical studies are necessary to fully assess its therapeutic potential.
Collapse
Affiliation(s)
- Chongyun Zhou
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Jiayun Wang
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Lili Zhou
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Hanxue Li
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Xing Liu
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Sen Wang
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Xingyu Zhang
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Xiaoqing Ye
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Hongyu Ren
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Kaile Zeng
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Xiuming Li
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Dan Wang
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China
| | - Jing Ji
- Jiangsu Key Laboratory for Screening of Marine Pharmaceutical Active Molecules, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222000, PR China; College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou, Jiangsu 225300, PR China.
| |
Collapse
|
64
|
Pang X, Zhao Y, Chen X, Wang M, Chen X, Yuan H, Sun Y, Han J, Zhao X. Preclinical Evaluation and First-in-Human Study of [ 68Ga]Ga-αvβ6-2: A Novel Dimeric Integrin αvβ6-Targeted PET Probe for Pancreatic Cancer Imaging. Mol Pharm 2025. [PMID: 40193102 DOI: 10.1021/acs.molpharmaceut.5c00051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Integrin αvβ6 is highly expressed in pancreatic cancer, making it an ideal target for molecular imaging diagnosis. Multimerization is considered an effective strategy to increase the accumulation of molecular probes in tumors. Here, we synthesized monomeric and dimeric αvβ6-targeting molecular probes, labeled with 68Ga, and designated them [68Ga]Ga-αvβ6-1 and [68Ga]Ga-αvβ6-2, respectively. Both in vitro and in vivo studies were conducted using human pancreatic cancer BxPC-3 cells and BxPC-3 tumor-bearing mice. Additionally, positron emission tomography/computed tomography (PET/CT) imaging with [68Ga]Ga-αvβ6-2 was performed in three patients with pancreatic cancer. In vitro studies demonstrated that [68Ga]Ga-αvβ6-2 exhibited greater binding affinity, cellular uptake, and internalization than did [68Ga]Ga-αvβ6-1. Micro-PET/CT imaging and biodistribution studies revealed the superior imaging performance of [68Ga]Ga-αvβ6-2. Furthermore, the first-in-human evaluation highlighted the favorable in vivo distribution and diagnostic efficacy of [68Ga]Ga-αvβ6-2 in pancreatic cancer. These results underscore the effectiveness of the multimerization strategy in the application of αvβ6-targeted molecular probes, suggesting that [68Ga]Ga-αvβ6-2 may possess favorable clinical translation potential.
Collapse
Affiliation(s)
- Xiao Pang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Medical Imaging College, North Sichuan Medical College; Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Yan Zhao
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Xiaolin Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Mengjiao Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Xiaoshan Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Huiqing Yuan
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Yuhan Sun
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Jingya Han
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei 050011, China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, Hebei 050011, China
| |
Collapse
|
65
|
Lin J, Lin S, Lin Z, Huang Z, Li D. Peroxynitrite as biomarker to evaluate the rehabilitation of cisplatin in the resistant cells with miR-125a-5p by using fluorescent assay. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 330:125663. [PMID: 39733707 DOI: 10.1016/j.saa.2024.125663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Reactive oxygen species (ROS) play a dual role in the chemotherapy of cancer with cisplatin, providing both anti-tumor effects and contributing to drug resistance at various stages of treatment which seriously affects treatment effectiveness. The detailed mechanism of ROS is urgently necessary to be explored. To address this issue in the non-small-cell lung cancer (NSCLC) with cisplatin-resistance, a reliable assay was developed by synthesizing and characterizing an interesting near-infrared (NIR) ONOO- probe BPB with high specificity, quick response (<30 s) and excellent limitation of detection (59 nM), which was further convinced through living cell imaging techniques providing different fluorescence variation between cell and cuvette. All the results revealed that ONOO- might be a practical biomarker to comprehend the detail molecular mechanism in cisplatin-resistant A549 cells. The elevated levels of ONOO- in cisplatin-resistant A549 cells, accompanied by a significant reduction in fluorescence following the knockout of miR-125a-5p in these cells and strong fluorescence without knockout of miR-125a-5p ignoring the presence of cisplatin. Comparing with cisplatin-resistant cells, the control would provide a rational background and then showed distinct fluorescence from BPB with ONOO- toward cisplatin. This assay offers a promising tool for exploring the molecular mechanisms associated with miR-125a-5p and its potential linkage to therapeutic efficacy involving ONOO- signaling. By utilizing this innovative assay, researchers can gain valuable insights into the treatment strategies and underlying mechanisms of cisplatin-resistant cancers, which should be beneficial to the therapy of cancers.
Collapse
Affiliation(s)
- Jinpei Lin
- Department of Integrative Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province 350014, China
| | - Shufang Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Zengyan Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Zhengrong Huang
- Department of Integrative Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province 350014, China.
| | - Daliang Li
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China.
| |
Collapse
|
66
|
Kozłowski M, Borzyszkowska D, Golara A, Lubikowski J, Cymbaluk-Płoska A. The Role of microRNA in the Prognosis and Diagnosis of Ovarian Cancer. Int J Mol Sci 2025; 26:3413. [PMID: 40244333 PMCID: PMC11989830 DOI: 10.3390/ijms26073413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
Ovarian cancer (OC) is one of the most common cancers in women. Biomarkers for OC are still being sought. The aim of this review was to evaluate microRNAs in the prognosis and diagnosis of OC. We conducted a literature review searching for articles published from January 2014 to September 2024. We included articles presenting the association of microRNAs with ovarian cancer prognosis, where patient survival was shown by the Kaplan-Meier curve, and articles presenting the association of microRNAs with ovarian cancer diagnosis, where the results were presented as an ROC curve. MicroRNAs are promising clinical markers in ovarian cancer patients. As is shown here, expression (high or low) of various miRNAs was differentially associated with survival in OC patients, with some miRNAs being associated with a longer survival and some with a shorter survival. In the absence of diagnostic markers for OC, the raised role of miRNAs in diagnosis seems all the more important. The diagnostic value of miRNAs has been shown, mostly as blood biomarkers, although they have also been evaluated as tissue or urine markers. MiRNAs have an important role as clinical biomarkers for ovarian cancer, not only as single molecules, but also as biomarker pairs or panels of miRNAs. It should be noted that most of the miRNAs reviewed here have been studied once, so despite the promising results, it seems necessary to conduct studies to confirm or negate the results obtained.
Collapse
Affiliation(s)
- Mateusz Kozłowski
- Department of Reconstructive Surgery and Gynecological Oncology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | | | | | | | | |
Collapse
|
67
|
Li Q, Pan P, Xian Q, Li J, Wang J, Cai J, Wang J, Jia Y, Sun H, Zhang L, Ma X. Alpha-estradiol and (R)-(-)-ibuprofen inhibit gastric cancer progression via GLI1 G-quadruplex. Front Pharmacol 2025; 16:1492694. [PMID: 40255562 PMCID: PMC12006100 DOI: 10.3389/fphar.2025.1492694] [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: 09/07/2024] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
Background The transcription factor GLI1, aberrantly activated in gastric cancer, drives tumor progression, yet no approved inhibitors currently target this molecule. G-quadruplex (G4) motifs in promoter regions have emerged as promising therapeutic targets. This study explores G4 stabilization in the GLI1 promoter as a novel strategy to suppress gastric cancer progression. Methods G4 formation in the GLI1 promoter was validated using circular dichroism. A dual-luciferase assay screened FDA-approved drugs for G4-stabilizing activity, identifying alpha-estradiol and (R)-(-)-ibuprofen as candidates. These compounds were evaluated for anti-tumor effects through in vitro assays (proliferation, migration, invasion) and in vivo xenograft models. Mechanistic insights into GLI1/PRKACB signaling were obtained via chromatin immunoprecipitation and pathway analysis. Results Stable G4 structures were confirmed in the GLI1 promoter. Alpha-estradiol and (R)-(-)-ibuprofen suppressed GLI1 transcription and protein levels, significantly inhibiting gastric cancer cell proliferation, migration, invasion, and stemness. In vivo, both compounds reduced tumor growth and metastasis, with (R)-(-)-ibuprofen synergizing with cisplatin to enhance efficacy. Mechanistically, GLI1 directly regulated PRKACB expression, and G4 stabilization downregulated PRKACB, impairing epithelial-mesenchymal transition and cancer stemness. Conclusion Targeting GLI1 G4 structures with alpha-estradiol and (R)-(-)-ibuprofen effectively inhibits gastric cancer progression by blocking GLI1/PRKACB signaling. This study highlights G4-targeted therapy as a novel and clinically translatable strategy for gastric cancer treatment.
Collapse
Affiliation(s)
- Qiang Li
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Pan Pan
- Department of Pathology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qingqing Xian
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jingtan Li
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jingting Wang
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jiaying Cai
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jing Wang
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yanfei Jia
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Haiji Sun
- College of Life Science, Shandong Normal University, Jinan, Shandong, China
| | - Lulu Zhang
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
68
|
Pang Y, Liang J, Deng Y, Chen W, Shen Y, Li J, Wang X, Ren Z. Identification and validation of HOXC6 as a diagnostic biomarker for Ewing sarcoma: insights from machine learning algorithms and in vitro experiments. Front Immunol 2025; 16:1449355. [PMID: 40255403 PMCID: PMC12006176 DOI: 10.3389/fimmu.2025.1449355] [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: 06/14/2024] [Accepted: 03/18/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction Early diagnosis of Ewing sarcoma (ES) is critical for improving patient prognosis. However, the accurate diagnosis of ES remains challenging, underscoring the need for novel diagnostic biomarkers to enhance diagnostic precision and reliability. This study aimed to identify potential gene expression-based biomarkers for the diagnosis of ES. Methods We selected the GSE17679, GSE45544, and GSE68776 datasets from the Gene Expression Omnibus (GEO) database. After correcting for batch effects, we combined ES and normal tissue samples from the GSE17679 and GSE45544 datasets to create a combined cohort. Two-thirds of both the tumor and normal samples from the combined cohort were randomly selected for the training cohort, while the remaining one-third served as the internal validation cohort. Additionally, the GSE68776 dataset was used for external validation. To identify key diagnostic genes, we applied three machine learning algorithms: least absolute shrinkage and selection operator (LASSO), support vector machine recursive feature elimination (SVM-RFE), and random forest (RF). Results HOXC6 was identified as a key diagnostic biomarker for ES. It demonstrated strong diagnostic performance across all cohorts, with area under the curve (AUC) values of 0.956 (95% CI: 0.909-0.990) in the training cohort, 0.995 (95% CI: 0.977-1.000) in the internal validation cohort, and 0.966 (95% CI: 0.910-0.999) in the external validation cohort. Functional validation through HOXC6 knockdown in the RD-ES cell line revealed that its suppression significantly inhibited cell proliferation and migration. Furthermore, transcriptome sequencing suggested potential oncogenic mechanisms underlying HOXC6 function. Discussion These findings highlight HOXC6 as a promising diagnostic biomarker for ES, demonstrating robust performance across multiple datasets. Additionally, its functional role suggests potential as a therapeutic target.
Collapse
Affiliation(s)
- Yonghua Pang
- Department of Orthopedics, The 904th Hospital of the Joint Logistics Support Force, People's Liberation Army of China, Wuxi, Jiangsu, China
| | - Jiahui Liang
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yakai Deng
- Department of Orthopedics, The 904th Hospital of the Joint Logistics Support Force, People's Liberation Army of China, Wuxi, Jiangsu, China
| | - Weinan Chen
- Department of Orthopedics, The 904th Hospital of the Joint Logistics Support Force, People's Liberation Army of China, Wuxi, Jiangsu, China
| | - Yunyan Shen
- Department of Orthopedics, The 904th Hospital of the Joint Logistics Support Force, People's Liberation Army of China, Wuxi, Jiangsu, China
| | - Jing Li
- Department of Orthopedics, Linyi People's Hospital, Linyi, Shandong, China
| | - Xin Wang
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Zhiyao Ren
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
69
|
Meyers JI, Schatz TM, Seitz CJ, Botbyl R, Moore BS, Crafts BG, Kitchen JR, Heaton S. Analytical Validation of MyProstateScore 2.0. Diagnostics (Basel) 2025; 15:923. [PMID: 40218273 PMCID: PMC11988380 DOI: 10.3390/diagnostics15070923] [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: 01/29/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Prostate cancer (PCa) is a leading cause of cancer-related deaths among men, with early detection playing a crucial role in improving outcomes. MyProstateScore 2.0 (MPS2), a novel urinary biomarker test, predicts clinically significant PCa to reduce invasive biopsy procedures. This study evaluates the analytical performance of MPS2 using both a post-digital rectal exam (DRE) and non-DRE urine samples. Methods: We assessed the reproducibility, precision, and detection limits of the eighteen MPS2 analytes. Analytical parameters including the linear range, upper and lower limits of quantification (ULOQ and LLOQ), and interference from substances commonly present in urine were evaluated. The reproducibility of the MPS2 scores was evaluated across post-DRE and non-DRE clinical urine samples. Results: MPS2 analytes demonstrated high linearity (R2 ≥ 0.975) across defined quantification ranges, with PCR efficiencies of 97-105%. The limits of detection (LOD) ranged from 40 to 160 copies/reaction, while the ULOQ was determined to be 106-107 copies/reaction for each analyte. Precision studies showed intra-run, inter-run, and inter-instrument standard deviations ≤0.5 Crt. Among the 12 potential interfering substances, only whole blood affected the performance of MPS2. The reproducibility of the MPS2 scores was consistent across post-DRE and non-DRE urine samples, meeting the acceptance criteria. Conclusions: The analytical validation confirms that MPS2 is robust and reliable in detecting biomarkers for clinically significant PCa. These findings, coupled with previous clinical validations, support the clinical use of MPS2 as a non-invasive diagnostic tool.
Collapse
Affiliation(s)
- Jacob I. Meyers
- Lynx Dx, Ann Arbor, MI 48108, USA; (T.M.S.); (C.J.S.); (R.B.); (B.S.M.); (B.G.C.); (J.R.K.); (S.H.)
| | | | | | | | | | | | | | | |
Collapse
|
70
|
Mao S, Li J, Huang J, Lv L, Zhang Q, Cheng Q, Liu X, Bi Z, Yao J. Therapeutic potential of microRNA-506 in cancer treatment: mechanisms and therapeutic implications. Front Oncol 2025; 15:1524763. [PMID: 40248198 PMCID: PMC12003368 DOI: 10.3389/fonc.2025.1524763] [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: 11/08/2024] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Cancer is a complex and highly lethal disease marked by unchecked cell proliferation, aggressive behavior, and a strong tendency to metastasize. Despite significant advancements in cancer diagnosis and treatment, challenges such as early detection difficulties, drug resistance, and adverse effects of radiotherapy or chemotherapy continue to threaten patient survival. MicroRNAs (miRNAs) have emerged as critical regulators in cancer biology, with miR-506 being extensively studied and recognized for its tumor-suppressive effects across multiple cancer types. This review examines the regulatory mechanisms of miR-506 in common cancers, focusing on its role in the competing endogenous RNA (ceRNA) network and its effects on cancer cell proliferation, apoptosis, and migration. We also discuss the potential of miR-506 as a therapeutic target and its role in overcoming drug resistance in cancer treatment. Overall, these insights underscore the therapeutic potential of miR-506 and its promise in developing novel cancer therapies.
Collapse
Affiliation(s)
- Shuzhen Mao
- Department of Pharmacy, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Junyan Li
- Department of Pathology, Second People’s Hospital of Ningyang, Taian, Shandong, China
| | - Jiahui Huang
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Lili Lv
- Department of Pathology, Second People’s Hospital of Ningyang, Taian, Shandong, China
| | - Qilian Zhang
- Department of Pathology, People’s Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qing Cheng
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Xiaojing Liu
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Zhiwei Bi
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Jing Yao
- Jining Key Laboratory of Pharmacology, School of Basic Medicine, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
71
|
Lin ZP, Gan G, Xu X, Wen C, Ding X, Chen XY, Zhang K, Guo WY, Lin M, Wang YY, Chen X, Xie C, Wang J, Li M, Zhong CQ. Comprehensive PTM profiling with SCASP-PTM uncovers mechanisms of p62 degradation and ALDOA-mediated tumor progression. Cell Rep 2025; 44:115500. [PMID: 40186868 DOI: 10.1016/j.celrep.2025.115500] [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: 10/29/2024] [Revised: 01/25/2025] [Accepted: 03/11/2025] [Indexed: 04/07/2025] Open
Abstract
Multiple post-translational modification (PTM) proteomics typically combines PTM enrichment with multiplex isobaric labeling and peptide fractionation. However, effective methods for sequentially enriching multiple PTMs from a single sample for data-independent acquisition mass spectrometry (DIA-MS) remain lacking. We present SDS-cyclodextrin-assisted sample preparation (SCASP)-PTM, an approach that enables desalting-free enrichment of diverse PTMs, including phosphopeptides, ubiquitinated peptides, acetylated peptides, glycopeptides, and biotinylated peptides. SCASP-PTM uses SDS for protein denaturation, which is sequestered by cyclodextrins before trypsin digestion, facilitating sequential PTM enrichment without additional purification steps. Combined with DIA-MS, SCASP-PTM quantifies the proteome, ubiquitinome, phosphoproteome, and glycoproteome in HeLa-S3 cell samples, identifying serine 28 phosphorylation as a key driver of poly(I:C)-induced p62 degradation. This method also quantifies PTMs in clinical tissue samples, revealing the critical role of ALDOA K330 ubiquitination/acetylation in tumor progression. SCASP-PTM offers a streamlined workflow for comprehensive PTM analysis in both basic research and clinical applications.
Collapse
Affiliation(s)
- Zhan-Peng Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Guohong Gan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiao Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Chengwen Wen
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xin Ding
- Department of Pathology, Zhongshan Hospital of Xiamen University, Xiamen University, Xiamen, Fujian 361004, China
| | - Xiang-Yu Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Kaijie Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Wen-Yu Guo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Mingxin Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yu-Yang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xi Chen
- SpecAlly Life Technology Co., Ltd., Wuhan, Hubei 430074, China
| | - Changchuan Xie
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jinling Wang
- Department of Emergency and Critical Care Center, The Second Affiliated Hospital of Guangdong Medical University, No. 12 Minyou Road, Xiashan, Zhanjiang, Guangdong 524003, China.
| | - Minjie Li
- Department of Thoracic Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian 361004, China.
| | - Chuan-Qi Zhong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
72
|
Yu Z, Cao W, Du C, Liu J, Peng L, Wei F. Developing a novel predictive model for identifying risk factors associated with being lost to follow-up among high-risk patients for recurrence following radical resection of hepatocellular carcinoma: the first report. BMC Cancer 2025; 25:597. [PMID: 40175947 PMCID: PMC11967030 DOI: 10.1186/s12885-025-14030-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 03/27/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Follow-up is essential especially for patients who are at a high risk of recurrence after radical resection of hepatocellular carcinoma (HCC). The aim of this study was to develop a predictive model aimed at identifying the risk factors associated with being lost to follow-up (LTFU) in high-risk patients for recurrence following radical resection of HCC. METHODS The retrospective study was conducted at our institution between October 2018 to May 2023. The patients who underwent radical liver resection for HCC and had high-risk factors for recurrence were categorized into an LTFU group and a control group. Multivariate logistic regression analysis was utilized to determine risk factors and construct a nomogram predictive model. RESULTS A total of 352 patients were included and subsequently classified into two distinct groups: the LTFU group (n = 123, 34.94%) and the control group (n = 229, 65.06%). Logistic regression analysis was then conducted to explore the potential associations between various factors and the occurrence of LTFU. The findings identified several independent risk factors for LTFU, including smoking (odds ratio, OR = 1.823, 95% confidence interval, CI 1.086-3.060, p = 0.023); residing more than 200 km away from the hospital (OR = 1.857, 95% CI 1.105-3.121, p = 0.019); having an unstable profession (OR = 1.918, 95% CI 1.112-3.311, p = 0.019); and lacking medical insurance (OR = 5.921, 95% CI 1.747-20.071, p = 0.004); the presence of liver cirrhosis (OR = 2.161, 95% CI 1.153-4.048, p = 0.016); an operation time less than 240 min (OR = 2.138, 95% CI 1.240-3.688, p = 0.006); and the absence of postoperative adjuvant therapy (OR = 2.641, 95% CI 1.504-4.637, p = 0.001). Based on these seven significant factors, a main effects model was established, designated as the Wei-LTFU model, which achieved an area under the curve value of 0.744 (95% CI 0.691-0.798) in predicting the likelihood of LTFU. CONCLUSION A main effects model, namely the Wei-LTFU model, incorporating the seven significant factors was formulated to predict the likelihood of LTFU occurrence, ultimately aiming to assist healthcare workers in developing effective strategies to improve follow-up outcomes for patients.
Collapse
Affiliation(s)
- Zichen Yu
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China
| | - Wenli Cao
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China
- Department of Public Health, Hangzhou Medical College, Hangzhou, 310059, Zhejiang Province, China
| | - Chengfei Du
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China
| | - Jie Liu
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China
| | - Liping Peng
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China
| | - Fangqiang Wei
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China.
| |
Collapse
|
73
|
Sato K, Takemura K, Oki R, Urasaki T, Yoneoka Y, Fujiwara R, Yasuda Y, Oguchi T, Numao N, Yamamoto S, Yonese J, Kume H, Yuasa T. Prognostic significance of body mass index in patients with metastatic renal cell carcinoma receiving first-line therapies. Urol Oncol 2025; 43:269.e1-269.e6. [PMID: 39757037 DOI: 10.1016/j.urolonc.2024.12.265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/02/2024] [Accepted: 12/15/2024] [Indexed: 01/07/2025]
Abstract
OBJECTIVES Higher body mass index (BMI) is reportedly associated with improved prognosis of patients with various cancers. However, it is unclear whether this phenomenon, also known as the obesity paradox, applies to metastatic renal cell carcinoma (mRCC). We aimed to determine the prognostic significance of BMI in patients with mRCC receiving first-line therapies. MATERIALS AND METHODS We retrospectively reviewed patients with mRCC receiving first-line immune checkpoint inhibitor (ICI)-based combination therapy or tyrosine kinase inhibitor monotherapy. Overall survival (OS) was defined as the time from systemic therapy initiation to death from any cause or last follow-up. Baseline patient characteristics were compared by Mann-Whitney U test or Fisher's exact test. OS curves were constructed by Kaplan-Meier estimates and were compared by log-rank test. Multivariable analysis was performed via Cox proportional-hazards regression. RESULTS Of the 183 patients included, 130 (71 %) were overweight (≥22 and 18 kg/m2 in men and women, respectively), and 63 (34 %) received ICI-based combination therapy. There was a significantly higher proportion of men in the overweight subgroup (87 % versus 64 %; P = 0.002). During the study period, 97 patients died, and median (95 % confidence interval) OS was 39.0 months (31.5-66.3 months) and 28.1 months (17.6-39.7 months) in overweight and normoweight patients, respectively (P = 0.015). On multivariable analysis, overweight was independently associated with longer OS (HR 0.57; P = 0.014). Subgroup analyses of patients receiving ICI-based combination therapy yielded similar results. CONCLUSION Overweight is associated with favorable outcomes in patients with mRCC receiving first-line therapies.
Collapse
Affiliation(s)
- Keigo Sato
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan; Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kosuke Takemura
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Ryosuke Oki
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tetsuya Urasaki
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yusuke Yoneoka
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryo Fujiwara
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yosuke Yasuda
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tomohiko Oguchi
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Noboru Numao
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shinya Yamamoto
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Junji Yonese
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Haruki Kume
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Yuasa
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
74
|
Zhou X, Yan S, Li D, Zhu H, Liu B, Liu S, Zhao W, Yang Z, Wu N, Li N. Radiolabelled anti-PD-L1 peptide PET/CT in predicting the efficacy of neoadjuvant immunotherapy combined with chemotherapy in resectable non-small cell lung cancer. Ann Nucl Med 2025; 39:364-372. [PMID: 39673015 DOI: 10.1007/s12149-024-02009-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND This study aimed to evaluate the predictive value of baseline PD-L1 targeted peptide 68Ga-NOTA-WL12 PET/CT in neoadjuvant immunotherapy combined with chemotherapy of resectable NSCLC. METHODS Patients with resectable NSCLC (n = 20) enrolled in this prospective study received baseline paired 68Ga-NOTA-WL12 PET/CT and 18F-FDG PET/CT. After 2-4 cycles of toripalimab plus nab-paclitaxel and cisplatin, surgery was performed if R0 resection was available. The major pathologic response (MPR) state of the post-operative specimen was recorded. The imaging parameters of the 68Ga-NOTA-WL12 PET/CT, 18F-FDG PET/CT and CT between the MPR and non-MPR groups and their predictive efficacy of MPR were compared. RESULTS Among 20 patients, 17 patients underwent surgery, 10 achieved an MPR and 7 did not. The SUVmax and tumour-to-blood pool (TBR) of baseline 68Ga-NOTA-WL12 in the MPR group were higher than those in the non-MPR group, and the difference in TBR was statistically significant. The ΔSULpeak% of 18F-FDG exhibited differences between the MPR and non-MPR groups with no significance. Baseline 18F-FDG PET/CT parameters and ΔD% failed to differentiate the two groups. The areas under the ROC curves of SUVmax, TBR in 68Ga-NOTA-WL12 PET/CT, ΔD% and ΔSULpeak% in 18F-FDG PET/CT were 0.76, 0.79, 0.71 and 0.80, respectively, in predicting MPR. CONCLUSION Baseline 68Ga-NOTA-WL12 PET/CT has a potential to predict the pathological response of neoadjuvant immunotherapy combined with chemotherapy in patients with resectable NSCLC, whose efficacy is comparable to that of therapy evaluations employing baseline and follow-up CT and 18F-FDG PET/CT examinations. TRIAL REGISTRATION NCT04304066, registered 13 November 2020, https://register. CLINICALTRIALS gov/prs/app/action/SelectProtocol?sid=S000AEI9&selectaction=Edit&uid=U000503E&ts=2&cx=-awajet .
Collapse
Affiliation(s)
- Xin Zhou
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Shi Yan
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Dan Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Hua Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Bing Liu
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Shiwei Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Wei Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China
| | - Zhi Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China.
| | - Nan Wu
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China.
| | - Nan Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Beijing, 100142, China.
| |
Collapse
|
75
|
Jadhav PA, Hole A, Saha P, Pansare K, Ghanwat A, Gera P, Bendale K, Krishna CM, Chaudhari P. Exploratory Raman Spectroscopic Studies of Canine Oral Tumour Types. JOURNAL OF BIOPHOTONICS 2025; 18:e202400372. [PMID: 39923801 DOI: 10.1002/jbio.202400372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 02/11/2025]
Abstract
Canine cancers are becoming increasingly significant due to their natural occurrence, similar to the spontaneous cancers in humans as well as their histological and biological similarities to human cancers. Several oral cancer types have been witnessed in dogs, based on the cell type from which the tumour originates. The type of oral tumour dictates severity of the disease, treatment options and prognoses. The current tissue-based Raman Spectroscopy (RS) study explores stratification of canine cancers. Raman spectra of histopathologically confirmed normal and oral tumour types namely Epulis, Spindle cell sarcoma (SCS), and Squamous cell carcinoma (SCC) were acquired using a Raman confocal microscope with 532 nm laser, pre-processed and multivariate analyses performed. PC-LDA achieved overall classification accuracy of > 70%. Thus, the present study evaluates potential of RS to identify the tumour type based on the identification of characteristic spectral features. Findings warrant large scale in vivo RS explorations in canine cancer subjects.
Collapse
Affiliation(s)
- Priyanka A Jadhav
- Chilakapati Laboratory, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - Arti Hole
- Chilakapati Laboratory, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | - Panchali Saha
- Chilakapati Laboratory, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - Kshama Pansare
- Chilakapati Laboratory, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | - Aishwarya Ghanwat
- Comparative Oncology Program & Small Animal Imaging Facility, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Poonam Gera
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
- Tissue Biorepository, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Kiran Bendale
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
- Comparative Oncology Program & Small Animal Imaging Facility, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - C Murali Krishna
- Chilakapati Laboratory, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - Pradip Chaudhari
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
- Comparative Oncology Program & Small Animal Imaging Facility, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| |
Collapse
|
76
|
Liu Y, Li C, Cui X, Li M, Liu S, Wang Z. Potentially diagnostic and prognostic roles of piRNAs/PIWIs in pancreatic cancer: A review. Biochim Biophys Acta Rev Cancer 2025; 1880:189286. [PMID: 39952623 DOI: 10.1016/j.bbcan.2025.189286] [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: 12/13/2024] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with limited early diagnostic methods and therapeutic options, contributing to its poor prognosis. Recent advances in high-throughput sequencing have highlighted the critical roles of noncoding RNAs (ncRNAs), particularly PIWI-interacting RNAs (piRNAs), in cancer biology. In this review, we systematically summarize the emerging roles of piRNAs and their associated PIWI proteins in PDAC pathogenesis, progression, and prognosis. We provide a comprehensive analysis of the molecular mechanisms by which piRNAs/PIWIs regulate gene expression and cellular signaling pathways in PDAC. Furthermore, we discuss their potential as novel biomarkers for early diagnosis and therapeutic targets. Importantly, this review identifies key piRNAs/PIWIs involved in PDAC and proposes innovative strategies for improving diagnosis and treatment outcomes. Our work not only consolidates current knowledge but also offers new perspectives for future research and clinical applications in PDAC management.
Collapse
Affiliation(s)
- Yukun Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changlei Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotong Cui
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Miaomiao Li
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China
| | - Shiguo Liu
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China.
| | - Zusen Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
77
|
Keys P, Corona K, McCown S, Huang L, Villarreal E, Chaij J, Tejera GDL, Tyler DS, Golovko G, Ayadi AE, Song J, Wolf SE. Impact of pre-existing cancer diagnoses on burn injury survival and morbidity. Burns 2025; 51:107414. [PMID: 39933421 DOI: 10.1016/j.burns.2025.107414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/27/2025] [Accepted: 02/02/2025] [Indexed: 02/13/2025]
Abstract
OBJECTIVES Severe burns place patients in a compromised state, especially those with pre-existing chronic diseases such as malignancy. Due to improvements in detection and treatment, cancer mortality has declined significantly, leaving a growing population of cancer survivors. Therefore, we wondered whether pre-existing cancer diagnoses influence patient outcomes following burn injury. METHODS 46,275 patients with a neoplasm diagnosis prior to experiencing a burn injury were identified using the TriNetX research network. Two control groups were organized: 1) cancer patients without burns and 2) burn patients with no history of cancer. The test group included patients with a pre-existing cancer diagnosis who received a subsequent burn injury. Outcomes included death, sepsis, nutritional deficiency, eating disorder, immunodeficiency, and depression. Odds ratios were calculated with 95 % confidence intervals. RESULTS Of the nearly 10 million cancer patients, 0.45 % experienced a burn injury after diagnosis. Compared to propensity-matched burned patients without cancer, burned patients with a previous cancer diagnosis had increased odds of developing sepsis (2.013, 1.895-2.138), nutritional deficiency (1.874, 1.58-2.221), immunodeficiency (5.584, 4.85-7.06), eating disorder (2.384, 1.947-2.918), and depression (1.772, 1.695-1.853). The odds ratios of sepsis (1.718, 1.612-1.83), nutritional deficiency (1.963, 1.593-2.418), immunodeficiency (1.265, 1.098-1.459), eating disorder (2.569, 2.077-3.177), and depression (1.538, 1.468-1.611) were increased when compared to cancer patients without burn injury. Burned patients with a previous neoplasm of hematologic origin fared worse in the odds of developing the mentioned outcomes compared to those with solid neoplasms. Lastly, burned patients with any previous neoplasm who later received chemotherapy and/or radiation suffered worse outcomes than those who never received such treatment. CONCLUSION Patients who developed neoplasms before receiving a burn injury demonstrated considerable increases in odds of poor outcomes. Increased odds of developing poor outcomes were greater in those with hematologic neoplasms and those who received chemotherapy or radiation treatment.
Collapse
Affiliation(s)
- Phillip Keys
- School of Medicine, University of Texas Medical Branch, Galveston, TX, United States.
| | - Kassandra Corona
- School of Medicine, University of Texas Medical Branch, Galveston, TX, United States.
| | - Sheldon McCown
- School of Medicine, University of Texas Medical Branch, Galveston, TX, United States.
| | - Lyndon Huang
- School of Medicine, University of Texas Medical Branch, Galveston, TX, United States.
| | - Elvia Villarreal
- School of Medicine, University of Texas Medical Branch, Galveston, TX, United States.
| | - Jasmine Chaij
- School of Medicine, University of Texas Medical Branch, Galveston, TX, United States.
| | - Giovanna De La Tejera
- School of Medicine, University of Texas Medical Branch, Galveston, TX, United States.
| | - Douglas S Tyler
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, United States.
| | - George Golovko
- Department of Pharmacology, University of Texas Medical Branch, Galveston, TX, United States.
| | - Amina El Ayadi
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, United States.
| | - Juquan Song
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, United States.
| | - Steven E Wolf
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, United States.
| |
Collapse
|
78
|
Duan Q, Li R, Wang M, Cui Z, Zhu X, Chen F, Han F, Ma J. Exploring the anti-NSCLC mechanism of phillyrin targeting inhibition of the HSP90-AKT pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3789-3802. [PMID: 39356318 DOI: 10.1007/s00210-024-03481-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 09/21/2024] [Indexed: 10/03/2024]
Abstract
Phillyrin (PHN), derived from the dried fruit of Forsythia suspensa (Thunb.) Vahl, is a kind of Chinese herbal medicine with the effect of clearing heat, and has been used in China for thousands of years in treating various tumors. However, the mechanism of its main components on non-small cell lung cancer (NSCLC) remains unclear. PHN is a distinct component extracted from Forsythia suspensa with promising anti-cancer activity against various tumor types. This study sought to elucidate the promising effects of PHN on NSCLC. Based on network pharmacology results, we identified potential PHN targets and pathways for NSCLC treatment. CCK-8 assay, wound healing assay, apoptosis assay, western blot, and in vivo experiments verified the inhibitory effect of PHN on NSCLC. Network pharmacology identified 160 potential PHN targets, 955 NSCLC-related targets, and 54 common targets, along with 132 pathways and 2 core genes. Biological experiments demonstrated that PHN significantly inhibited the growth and migration of A549 and LLC cells while promoting their apoptosis. Western blot analysis revealed down-regulation of AKT, HSP90AA1, and CDC37 expression, suggesting that PHN inhibits A549 and LLC cell proliferation by down-regulating the HSP90-AKT pathway. In vivo experiments confirmed that PHN significantly inhibited NSCLC growth with low toxicity. This study, using network pharmacology and biological experiments, verified the effectiveness of PHN against NSCLC through the HSP90-AKT pathway. These findings provide a foundation for further research and analysis.
Collapse
Affiliation(s)
- Qiong Duan
- The Affiliated Lianyungang Municipal Oriental Hospital of Xuzhou Medical University, Lianyungang, 222042, China
| | - Ruochen Li
- Sichuan Integrative Medicine Hospital, Chengdu, 610000, China
| | - Mingxiao Wang
- Sichuan Integrative Medicine Hospital, Chengdu, 610000, China
| | - Zhenting Cui
- The Affiliated Lianyungang Municipal Oriental Hospital of Xuzhou Medical University, Lianyungang, 222042, China
| | - Xia Zhu
- The Affiliated Lianyungang Municipal Oriental Hospital of Xuzhou Medical University, Lianyungang, 222042, China
| | - Fanghong Chen
- The Affiliated Lianyungang Municipal Oriental Hospital of Xuzhou Medical University, Lianyungang, 222042, China
| | - Feng Han
- The Affiliated Lianyungang Municipal Oriental Hospital of Xuzhou Medical University, Lianyungang, 222042, China.
| | - Jianxin Ma
- The Affiliated Lianyungang Municipal Oriental Hospital of Xuzhou Medical University, Lianyungang, 222042, China.
| |
Collapse
|
79
|
Tao Q, Li Y, Zhang W, Zhang M, Li X, Jin H, Zheng J, Li Y. Long non-coding RNA ZFAS1 promotes ferroptosis by regulating the miR-185-5p/SLC25A28 axis in clear cell renal cell carcinoma. Int J Biol Macromol 2025; 304:140602. [PMID: 39922352 DOI: 10.1016/j.ijbiomac.2025.140602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/04/2024] [Accepted: 11/11/2024] [Indexed: 02/10/2025]
Abstract
Ferroptosis is a novel, iron-dependent regulated cell death mode. The biochemical features of ferroptosis include iron accumulation, lipid peroxidation, inhibition of glutathione peroxidase 4 (GPX4) and antioxidant glutathione (GSH) decrease through inhibition of the system xc- transporter. Zinc finger NFX1 type-containing 1 (ZNFX1) antisense RNA 1 (ZFAS1) is a long non-coding RNA that has been identified as an oncogene in various types of cancers. However, its regulatory role and molecular mechanisms in clear cell renal cell carcinoma (ccRCC) ferroptosis remain unclear. In this study, the ferroptosis inducers (FINS) (erastin and RSL3) were found to increase ZFAS1 expression through the facilitation of SP1 binding to the ZFAS1 promoter. ZFAS1 increased mRNA and protein levels of solute carrier family 25 member 28 (SLC25A28) via functioning as a miR-185-5p sponge. Overexpressed SLC25A28 increased the production of ROS and caused a decrease in NADPH and GSH in cells treated with FINS. In addition, overexpression of ZFAS1 enhanced ferroptosis both in vitro and in vivo. Altogether, this study demonstrates that ZFAS1 is a crucial element of ferroptosis in ccRCC, as it is responsible for the regulation of miR-185-5p and SLC25A28. Introducing ferroptosis could be a beneficial approach to treat ccRCC patients with high ZFAS1 levels.
Collapse
Affiliation(s)
- Qiqi Tao
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yifei Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weizhi Zhang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Minghong Zhang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinmiao Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hui Jin
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianjian Zheng
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Yeping Li
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
80
|
Meng X, Yang D, Jin H, Xu H, Lu J, Liu Z, Wang Z, Wang L, Yang Z. MRI-based radiomics model for predicting endometrial cancer with high tumor mutation burden. Abdom Radiol (NY) 2025; 50:1822-1830. [PMID: 39417854 DOI: 10.1007/s00261-024-04547-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 10/19/2024]
Abstract
PURPOSE To evaluate the performance of MRI-based radiomics in predicting endometrial cancer (EC) with a high tumor mutation burden (TMB-H). METHODS A total of 122 patients with pathologically confirmed EC (40 TMB-H, 82 non-TMB-H) were included in this retrospective study. Patients were randomly divided into training and testing cohorts in a ratio of 7:3. Radiomics features were extracted from sagittal T2-weighted images and contrast-enhanced T1-weighted images. Then, the logistic regression (LR), random forest (RF), and support vector machine (SVM) algorithms were used to construct radiomics models. The area under the receiver operating characteristic curve (AUC) was calculated to evaluate the diagnostic performance of each model, and decision curve analysis was used to determine their clinical application value. RESULTS Four radiomics features were selected to build the radiomics models. The three models had similar performance, achieving 0.771 (LR), 0.892 (RF), and 0.738 (SVM) in the training cohort, and 0.787 (LR), 0.798 (RF), and 0.777 (SVM) in the testing cohort. The decision curve demonstrated the good clinical application value of the LR model. CONCLUSIONS The MRI-based radiomics models demonstrated moderate predictive ability for TMB-H EC and thus may be a tool for preoperative, noninvasive prediction of TMB-H EC.
Collapse
Affiliation(s)
- Xuxu Meng
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Dawei Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - He Jin
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hui Xu
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jun Lu
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhenhao Liu
- Department of Radiology, Affiliated Hospital of Changzhi Institute of Traditional Chinese Medicine, Changzhi, China
| | - Zhenchang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Liang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Zhenghan Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
81
|
Lin CY, Fang JY, Hsiao CY, Lee CW, Alshetaili A, Lin ZC. Dual cell-penetrating peptide-conjugated polymeric nanocarriers for miRNA-205-5p delivery in gene therapy of cutaneous squamous cell carcinoma. Acta Biomater 2025; 196:332-349. [PMID: 40015353 DOI: 10.1016/j.actbio.2025.02.056] [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: 10/13/2024] [Revised: 02/16/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Despite the potential of microRNAs (miRNAs) in suppressing tumorigenesis, the main challenges are achieving tumor-specific selectivity and efficient delivery into cancer cells. In this study, miR-205-5p-loaded polymeric nanoparticles conjugated with dual cell-penetrating peptides (CPPs) were designed for targeting and treating cutaneous squamous cell carcinoma (cSCC). The CPPs, R9, and p28, demonstrated high cell-penetrating/targeting abilities and antitumor activity. The anti-cSCC effect of the nanocarriers was examined using in vitro cellular 2D and 3D models and in vivo spheroid-xenografted murine models. The average size of the dual CPP-conjugated nanocarriers was 193 nm with a zeta potential of 5.7 mV. These nanocarriers were readily internalized by A431 cells, resulting in decreased proliferation compared to naked agomiR and nanoparticles with a single CPP. The nanocarriers induced cell cycle arrest in the G0/G1 stage. By loading the miR-205-5p mimic, the dual CPP-conjugated nanoparticles enhanced cell apoptosis threefold compared to the control, activating caspases and poly(ADP-ribose) polymerase (PARP). The wound healing assay demonstrated that the nanocarriers significantly inhibited the migration and invasion of cSCC cells. Additionally, the CPP-conjugated nanocarriers penetrated cSCC 3D spheroids, reducing spheroidal size and proliferation. In vivo studies demonstrated that the intratumoral CPP-conjugated nanocarriers achieved a 30 % reduction in tumor volume than the PBS control. The number of Ki67-positive cells in the nanocarrier-treated tumor decreased fivefold than the untreated tumors. The nanoparticulate agomiR (1 μM) exhibited no cytotoxicity towards normal keratinocytes. No significant toxicity was observed in the skin and peripheral organs following subcutaneous administration of the nanoparticles in healthy mice. These findings demonstrate that miR-205-5p mimic delivery via dual CPP-conjugated nanocarriers can promote efficient and safe cSCC regression. STATEMENT OF SIGNIFICANCE: Cutaneous squamous cell carcinoma (cSCC) is a highly invasive skin malignancy with limited treatment options. This study introduces dual cell-penetrating peptide (CPP)-conjugated polymeric nanoparticles for delivering miR-205-5p, a tumor-suppressor microRNA, to cSCC cells. The nanosystem enhances cellular uptake, inhibits cell proliferation, and promotes apoptosis in both 2D and 3D tumor models. In vivo, the nanocarriers demonstrate significant antitumor efficacy with minimal toxicity, highlighting their potential as a targeted, non-invasive therapy. This research represents a promising advance in gene therapy for cSCC by combining nanotechnology and CPPs to address challenges in miRNA delivery and tumor targeting.
Collapse
Affiliation(s)
- Cheng-Yu Lin
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan; Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chien-Yu Hsiao
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan; Department of Nutrition and Health Sciences, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan; Aesthetic Medical Center, Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chiang-Wen Lee
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan; Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi, Chiayi, Taiwan; Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi, Chiayi, Taiwan; Department of Respiratory Care, Chang Gung University of Science and Technology, Puzi, Chiayi, Taiwan
| | - Abdullah Alshetaili
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Zih-Chan Lin
- Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi, Chiayi, Taiwan.
| |
Collapse
|
82
|
Adler Jaffe S, Kano M, Rieder S, Gundelach AC, Boyce T, Rutledge T, Dayao Z, Sussman AL. "Care needs to be integrated" Patient and provider perspectives on a cancer shared-care model. J Cancer Surviv 2025; 19:526-533. [PMID: 37889397 DOI: 10.1007/s11764-023-01486-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 10/15/2023] [Indexed: 10/28/2023]
Abstract
PURPOSE Current early-stage breast and gynecological cancer care models often begin with a referral from a primary care provider (PCP) or gynecologist (OB/Gyn) and end with a patient being transitioned back to the referring provider at the completion of treatment. There is frequently little communication between oncologists and the referring provider during treatment, and this pattern continues after the patient completes their treatment. METHODS We convened a diverse Patient Advisory Board (PAB) to identify areas where breast or gynecological cancer patients felt they could benefit from additional support during and after their cancer care. PAB members attended five Zoom meetings and completed four online surveys. Semi-structured interviews were conducted with primary care or OB/Gyn physicians to collect information on current practices. RESULTS Patients identified multiple areas in which they needed additional support from their PCP. Providers also identified topics on which they could use additional training. However, there was little overlap between patient and provider priority topics. Both patients and providers agreed that there was inadequate communication between the cancer center and PCPs before, during, and after cancer treatment. CONCLUSIONS A shared-care model that emphasizes communication between primary care providers, the oncology care team, and patients is urgently needed. Patients indicated the need for additional support from their PCP on specific topics, and PCPs were interested in continuing their education to better serve their patients with cancer. IMPLICATIONS FOR CANCER SURVIVORS The importance of consistent communication among all parties during the entire cancer journey was emphasized as a key area for improvement.
Collapse
Affiliation(s)
| | - Miria Kano
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Stephanie Rieder
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
- Department of Obstetrics and Gynecology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Amy C Gundelach
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Tawny Boyce
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Teresa Rutledge
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
- Department of Obstetrics and Gynecology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Zoneddy Dayao
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Andrew L Sussman
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
- Department of Family and Community Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
83
|
Najafi A, Heidary M, Martinez RM, Baby AR, Morowvat MH. Microalgae-based sunscreens as green and sustainable cosmetic products. Int J Cosmet Sci 2025; 47:213-222. [PMID: 39295125 DOI: 10.1111/ics.13019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/27/2024] [Accepted: 08/13/2024] [Indexed: 09/21/2024]
Abstract
Recently, microalgal biotechnology has attained great acceptance among various researchers and industries for the green and sustainable production of different bioactive compounds. They provide multiple metabolites and molecules, making them an ideal candidate for cosmetic formulators and cosmeceutical companies. Nevertheless, numerous microalgae strains have never been studied for their pharmaceutical, nutritional and cosmeceutical purposes. Even less, only some have been cultivated on a large scale for bioactive compound production. Here, we have studied the cosmetic and cosmeceutical potentials of different microalgal strains for sunscreen as adjuvants and boosters in a green, carbon-neutral and sustainable platform. Other bioactive compounds were exploited, and the available products in the market and the published patents were also reviewed. From our review, it will be possible to combine the fundamental and practical aspects of microalgal biotechnology toward a greener and more sustainable future for the cosmetic/cosmeceutical areas of the photoprotection scenario.
Collapse
Affiliation(s)
- Asal Najafi
- Department of Pharmacology and Toxicology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Heidary
- Department of Pharmacology and Toxicology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Renata Miliani Martinez
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - André Rolim Baby
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Mohammad Hossein Morowvat
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
84
|
Sands J, Ahn MJ, Lisberg A, Cho BC, Blumenschein G, Shum E, Pons Tostivint E, Goto Y, Yoh K, Heist R, Shimizu J, Lee JS, Baas P, Planchard D, Pérol M, Felip E, Su WC, Zebger-Gong H, Lan L, Liu C, Howarth P, Chiaverelli R, Paz-Ares L. Datopotamab Deruxtecan in Advanced or Metastatic Non-Small Cell Lung Cancer With Actionable Genomic Alterations: Results From the Phase II TROPION-Lung05 Study. J Clin Oncol 2025; 43:1254-1265. [PMID: 39761483 PMCID: PMC11949215 DOI: 10.1200/jco-24-01349] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/05/2024] [Accepted: 11/14/2024] [Indexed: 03/29/2025] Open
Abstract
PURPOSE Datopotamab deruxtecan (Dato-DXd) is a trophoblast cell-surface antigen-2-directed antibody-drug conjugate with a highly potent topoisomerase I inhibitor payload. The TROPION-Lung05 phase II trial (ClinicalTrials.gov identifier: NCT04484142) evaluated the safety and clinical activity of Dato-DXd in patients with advanced/metastatic non-small cell lung cancer (NSCLC) with actionable genomic alterations progressing on or after targeted therapy and platinum-based chemotherapy. PATIENTS AND METHODS Patients received Dato-DXd 6 mg/kg once every 3 weeks. The primary end point was objective response rate (ORR) by blinded independent central review. Secondary end points included duration of response (DOR), safety, tolerability, and survival. RESULTS Among 137 patients who received at least 1 dose of Dato-DXd, 71.5% received at least three lines of prior therapies for advanced/metastatic disease. Overall, 56.9% had EGFR mutations and 24.8% had ALK rearrangements. Median treatment duration was 4.4 months (range, 0.7-20.6). The confirmed ORR was 35.8% (95% CI, 27.8 to 44.4) overall, and 43.6% (95% CI, 32.4 to 55.3) and 23.5% (95% CI, 10.7 to 41.2) in those with EGFR mutations and ALK rearrangements, respectively. The median DOR was 7.0 months (95% CI, 4.2 to 9.8), and the overall disease control rate was 78.8% (95% CI, 71.0 to 85.3). Grade ≥3 treatment-related adverse events (TRAEs) occurred in 28.5% of patients. The most common TRAE was stomatitis (preferred term; any grade: 56.2%; grade ≥3: 9.5%). Five (3.6%) patients experienced adjudicated treatment-related interstitial lung disease/pneumonitis, with 1 (0.7%) grade 5 event. CONCLUSION Encouraging and durable antitumor activity was observed with Dato-DXd in this heavily pretreated advanced/metastatic NSCLC population with actionable genomic alterations. The rate of treatment-related grade ≥3 toxicities was comparable with previous observations, and no new safety signals were observed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Kiyotaka Yoh
- National Cancer Center Hospital East, Kashiwa, Japan
| | - Rebecca Heist
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Harvard University, Boston, MA
| | | | - Jong-Seok Lee
- Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Paul Baas
- Dana-Farber Cancer Institute, Boston, MA
| | | | | | | | - Wu-Chou Su
- Dana-Farber Cancer Institute, Boston, MA
| | | | - Lan Lan
- Daiichi Sankyo, Inc, Basking Ridge, NJ
| | | | | | | | | |
Collapse
|
85
|
Tan J, Zhao D, Wang Q, Peng Y, Li J, Li X, Che N, Hu Y, Zheng H. Whole Exome Sequencing Study Identifies Distinct Characteristics of Transformed Small Cell Lung Cancer With EGFR Mutation Compared to De Novo Small Cell and Primary Non-Small Cell Lung Cancers. Cancer Med 2025; 14:e70838. [PMID: 40197849 PMCID: PMC11976457 DOI: 10.1002/cam4.70838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR)-mutated lung adenocarcinoma (LUAD) is the most common subtype among non-small cell lung cancer (NSCLC) and targeted therapies are the primary approach for treatment. However, the development of resistance to therapy and histological transformation into small cell lung cancer (SCLC) present significant challenges. Understanding the mechanisms underlying this transformation is crucial for effective differential diagnosis and the formulation of treatment strategies. METHODS In this study, we collected tissue from 5 primary LUAD before SCLC transformation, 12 transformed SCLC after EGFR tyrosine kinase inhibitor (TKI) treatment, and 18 de novo SCLC from lung cancer patients treated at Beijing Chest Hospital, Capital Medical University from January 2015 to December 2021. Whole-exome sequencing was performed on these samples to compare the genomic alterations of these three tumor types, elucidating their similarities, differences, and connections. Statistical analyses were conducted using the Fisher exact test and performed with R v4.2.1 environment. RESULTS Among 12 transformed SCLC cases, the majority were female (10/12, 83.3%), non-smokers (10/12, 83.3%) and harbored EGFR 19del mutations (11/12, 91.7%). Four were with limited stage and 8 with extensive stage. TP53 mutations and RB1 loss are important but not necessary for SCLC transformation. The mutation rates of TP53 were 60% (3/5) in primary LUAD, 70% (7/10) in transformed SCLC, and 89% (16/18) in de novo SCLC. RB1 loss rates were 40% (2/5) in primary LUAD, 30% (3/10) in transformed SCLC, and 50% (9/18) in de novo SCLC. Additionally, mutations in COL22A1 and ALMS1 were only observed in transformed SCLC and de novo SCLC. In contrast, mutations in PTCH2, CNGB3, SPTBN5, CROCC, and MYO15A were more common in transformed SCLC, whereas PABPC3 and MUC19 mutations were more frequent in de novo SCLC. Smoking-related mutations (SBS4) were only found in de novo SCLC, with no changes observed in transformed SCLC. TMB levels were significantly lower in transformed SCLC compared to de novo SCLC (p = 0.01). Genomic instability was significantly higher in transformed SCLC compared to primary LUAD and de novo SCLC. This was supported by higher levels of homologous recombination deficiency (HRD, p = 0.025), uniparental disomy (UPD, p = 0.003), loss of heterozygosity (LOH, p = 0.008), and telomeric allelic imbalance (TAI, p = 0.02). The increased frequency of UPD events in transformed SCLC suggests that UPD may act as a "second hit" in Knudson's model, leading to biallelic inactivation of tumor suppressor genes. High similarity was observed in genetic alterations related to DNA damage repair (DDR) and Notch signaling pathways between transformed SCLC and de novo SCLC. CONCLUSIONS The identification of these specific genomic alterations in transformed SCLC contributes to a better understanding of the mechanisms driving this transformation. This knowledge may guide future predicting the transformation of SCLC and the development of personalized treatment strategies for these patients.
Collapse
Affiliation(s)
- Jinjing Tan
- Cancer Research Center, Beijing Chest HospitalCapital Medical University, Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingChina
| | - Dan Zhao
- Department of Pathology, Beijing Chest HospitalCapital Medical University, Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingChina
| | - Qunhui Wang
- Department of Medical Oncology, Beijing Chest HospitalCapital Medical University, Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingChina
| | - Yanjing Peng
- Cancer Research Center, Beijing Chest HospitalCapital Medical University, Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingChina
| | - Jie Li
- Department of Medical Oncology, Beijing Chest HospitalCapital Medical University, Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingChina
| | - Xi Li
- Department of Medical Oncology, Beijing Chest HospitalCapital Medical University, Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingChina
| | - Nanying Che
- Department of Pathology, Beijing Chest HospitalCapital Medical University, Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingChina
| | - Ying Hu
- Department of Medical Oncology, Beijing Chest HospitalCapital Medical University, Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingChina
| | - Hua Zheng
- Department of Medical Oncology, Beijing Chest HospitalCapital Medical University, Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingChina
| |
Collapse
|
86
|
Devasahayam Arokia Balaya R, Sen P, Grant CW, Zenka R, Sappani M, Lakshmanan J, Athreya AP, Kandasamy RK, Pandey A, Byeon SK. An integrative multi-omics analysis reveals a multi-analyte signature of pancreatic ductal adenocarcinoma in serum. J Gastroenterol 2025; 60:496-511. [PMID: 39666045 DOI: 10.1007/s00535-024-02197-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 12/01/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains a formidable health challenge due to its detection at a late stage and a lack of reliable biomarkers for early detection. Although levels of carbohydrate antigen 19-9 are often used in conjunction with imaging-based tests to aid in the diagnosis of PDAC, there is still a need for more sensitive and specific biomarkers for early detection of PDAC. METHODS We obtained serum samples from 88 subjects (patients with PDAC (n = 58) and controls (n = 30)). We carried out a multi-omics analysis to measure cytokines and related proteins using proximity extension technology and lipidomics and metabolomics using tandem mass spectrometry. Statistical analysis was carried out to find molecular alterations in patients with PDAC and a machine learning model was used to derive a molecular signature of PDAC. RESULTS We quantified 1,462 circulatory proteins along with 873 lipids and 1,001 metabolites. A total of 505 proteins, 186 metabolites and 33 lipids including bone marrow stromal antigen 2 (BST2), keratin 18 (KRT18), and cholesteryl ester(20:5) were found to be significantly altered in patients. We identified different levels of sphingosine, sphinganine, urobilinogen and lactose indicating that glycosphingolipid and galactose metabolisms were significantly altered in patients compared to controls. In addition, elevated levels of diacylglycerols and decreased cholesteryl esters were observed in patients. Using a machine learning model, we identified a signature of 38 biomarkers for PDAC, composed of 21 proteins, 4 lipids, and 13 metabolites. CONCLUSIONS Overall, this study identified several proteins, metabolites and lipids involved in various pathways including cholesterol and lipid metabolism to be changing in patients. In addition, we discovered a multi-analyte signature that could be further tested for detection of PDAC.
Collapse
Affiliation(s)
| | - Partho Sen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Caroline W Grant
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Roman Zenka
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Marimuthu Sappani
- Department of Biostatistics, Christian Medical College, Vellore, Tamil Nadu, 632002, India
| | - Jeyaseelan Lakshmanan
- College of Medicine, Mohammad Bin Rashid University of Medicine and Health Sciences, Dubai, 505055, UAE
| | - Arjun P Athreya
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Richard K Kandasamy
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
- Manipal Academy of Higher Education, Manipal, Karnataka, 5761904, India
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
- Manipal Academy of Higher Education, Manipal, Karnataka, 5761904, India.
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
87
|
Tseng SH, Wang KM, Su TY, Wang KJ. Survivability prognosis of lung cancer patients with comorbidities-a Gaussian Bayesian network model. Med Biol Eng Comput 2025; 63:1201-1213. [PMID: 39695068 DOI: 10.1007/s11517-024-03261-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 11/25/2024] [Indexed: 12/20/2024]
Abstract
Comorbidities are influencing factors that cause lung cancer. An accurate survivability prediction model is required considering these confounding factors (a variety of comorbidities and treatments). The study developed a conditional Gaussian Bayesian network (CGBN) model to predict the related survival time with likelihood under various conditions. The lung cancer patients were collected from the National Health Insurance Research Database in Taiwan. Six major chronic diseases (i.e., pulmonary tuberculosis, COPD, kidney failure, diabetes mellitus, stroke, and liver disease) are investigated. A total of 2875 lung cancer cases with key comorbidities were selected. This study examined three types of lung cancer treatment: surgery, chemotherapy, and targeted therapy. The study outcomes provided the likelihood of survival time occurrences. Survival analysis indicates that diabetes mellitus and liver disease are significantly riskier than the other comorbidities for lung cancer patients. The proposed CGBN model achieved high accuracy as compared to the existing literature. The proposed CGBN model is advantageous for modeling the relationship between numerical and categorical influencing factors and response variables for lung cancer with comorbidities. The proposed model facilitates the flexible and accurate estimation of various lung cancer-related queries.
Collapse
Affiliation(s)
- Shih-Hsien Tseng
- Department of Industrial Management, National Taiwan University of Science and Technology (NTUST), No.43, Sec. 4, Keelung Rd., Da'an Dist., Taipei, 106, Taiwan, ROC
| | - Kung-Min Wang
- Department of Industrial Management, National Taiwan University of Science and Technology (NTUST), No.43, Sec. 4, Keelung Rd., Da'an Dist., Taipei, 106, Taiwan, ROC
- Department of Surgery, Shin-Kong Wu Ho-Su Memorial Hospital, Shilin District, Taipei, 111, Taiwan, ROC
| | - Ting-Yang Su
- Department of Industrial Management, National Taiwan University of Science and Technology (NTUST), No.43, Sec. 4, Keelung Rd., Da'an Dist., Taipei, 106, Taiwan, ROC
| | - Kung-Jeng Wang
- Department of Industrial Management, National Taiwan University of Science and Technology (NTUST), No.43, Sec. 4, Keelung Rd., Da'an Dist., Taipei, 106, Taiwan, ROC.
| |
Collapse
|
88
|
Zi CT, Wu YL, Liu ZH, Niu Y, Yuan WJ, Yang ZW, Wang XJ, Sun XL, Yang L, Sheng J. Novel (-)-eigallocatechin-3-gallate-erlotinib conjugates via triazole rings inhibit non-small cell lung cancer cells through EGFR signaling pathway. Bioorg Chem 2025; 157:108263. [PMID: 39938444 DOI: 10.1016/j.bioorg.2025.108263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/19/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
EGFR is frequently overexpressed in non-small cell lung cancer, and EGFR plays a crucial role in the occurrence and progression of malignant tumors. Currently, drug resistance often develops following treatment with EGFR tyrosine kinase inhibitors, such as erlotinib and gefitinib. Therefore, It is essential to investigate new compounds that can effectively target EGFR overexpression. The polyphenols epigallocatechin-3-gallate (EGCG), found in tea, have demonstrated anti-cancer properties. In this study, we linked EGCG and erlotinib through a click reaction using polyglycol to form an EGCG-erlotinib conjugated compounds (EGCG-Erls). We then explored its biological activity through various experiments. The results indicated that the compound 10 exhibited a superior inhibitory effect on NCI-H1975 cells, reduced their cloning and migratory capabilities, promoted cell apoptosis, and inhibited cell cycle progression. Furthermore, it was observed that compound 10 can bind to the EGFR protein and effectively inhibit the expression of phosphorylated EGFR (p-EGFR) and its downstream signaling proteins. Overall, the study suggests that compound 10 may induce apoptosis and inhibit cell proliferation via the EGFR signaling pathway, providing a promising avenue for the development of new EGFR inhibitors.
Collapse
Affiliation(s)
- Cheng-Ting Zi
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China; Institute of Biofabrication Research, Yunnan Agricultural University, College of Science, Kunming 650201, China
| | - Yi-Long Wu
- Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China; Institute of Biofabrication Research, Yunnan Agricultural University, College of Science, Kunming 650201, China
| | - Zhen-Hao Liu
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Yun Niu
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Wen-Juan Yuan
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China; Institute of Biofabrication Research, Yunnan Agricultural University, College of Science, Kunming 650201, China
| | - Zi-Wei Yang
- Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China; Institute of Biofabrication Research, Yunnan Agricultural University, College of Science, Kunming 650201, China
| | - Xuan-Jun Wang
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Xiu-Li Sun
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| | - Liu Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
| | - Jun Sheng
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| |
Collapse
|
89
|
Zhang J, Deng Y, Tang H, Liu J, Si W, Sun B, Zhuang K. New predictors of recurrence in post-polypectomy surveillance - A retrospective analysis of risk stratification for advanced adenomas and polyps. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2025; 117:188-197. [PMID: 39559908 DOI: 10.17235/reed.2024.10832/2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
BACKGROUND AND AIMS recent guidelines emphasized that not all patients with recurrent polyps benefit from post-polypectomy surveillance, except for advanced adenomas recurrence. This study aimed to analyze the recurrence risk factors for advanced adenomas and investigated the difference from any polyp recurrence. METHODS this retrospective observational study included patients who underwent colonoscopy and at least one post-polypectomy surveillance. Multivariate regression models identified risk factors for the recurrence of polyps and advanced adenomas. The Youden index determined the optimal cut-off value for risk factors. Kaplan-Meier curve estimated the cumulative recurrence rates, and the log-rank tests compared the differences between these curves. RESULTS a total of 1,818 patients had polyps at baseline examination. During post-polypectomy surveillance, 1,063 patients had recurrent polyps, and 64 patients experienced recurrent advanced adenomas. Multivariate logistic analysis identified age as an independent risk factor for both advanced adenoma (OR [95 % CI]: 1.028 [1.003-1.056]) and polyps (OR [95 % CI]: 1.019 [1.009-1.028]), with cut-off values of 57 years and 53 years, respectively. A cut-off value of 40 years can increase the predictive sensitivity to 95 %. Polyps size (OR [95 % CI]: 1.070 [1.014-1.147]) and high-risk pathology (OR [95 % CI]: 6.339 [2.057-23.919]) were significantly associated with recurrence of advanced adenomas, but not with any polyps, with a size cut-off value of 7.5 mm. Except for high-risk pathology, neither hyperplastic polyps nor tubular adenomas with low-grade dysplasia increased the recurrence risk of advanced adenomas. CONCLUSIONS a cut-off value of 40 years can increase the predictive sensitivity to 95 % for both advanced adenomas and any polyps. Polyps size and high-risk pathology were associated with the recurrence of advanced adenoma, but not with any polyps.
Collapse
Affiliation(s)
- Jun Zhang
- Gastroenterology and Hepatology, Xi'an Central Hospital
| | - You Deng
- Liver Research Center, Beijing Friendship Hospital
| | - Hailing Tang
- Gastroenterology and Hepatology, Xi'an Central Hospital
| | - Jiaming Liu
- Gastroenterology and Hepatology, Xi'an Central Hospital
| | - Wangli Si
- Gastroenterology and Hepatology, Xi'an Central Hospital
| | - Baihe Sun
- Gastroenterology and Hepatology, Xi'an Central Hospital
| | - Kun Zhuang
- Gastroenterology and Hepatology, Xi'an Central Hospital, China
| |
Collapse
|
90
|
Ingaldi C, Minghetti M, D'Ambra V, Ricci C, Alberici L, Casadei R. Pancreatic cancer resection in the elderly: state of the art, and future challenges. A systematic review. Minerva Surg 2025; 80:138-149. [PMID: 40261182 DOI: 10.23736/s2724-5691.25.10527-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
INTRODUCTION Pancreatic cancer resection in the elderly population represents a challenging problem. In addition, a chronological age for the "elderly" is lacking. The aim of the present review was to assess the safety and feasibility of major pancreatic tumor resection in elderly patients, considering different age cut-offs (≥70; ≥75 and ≥80 years old). EVIDENCE ACQUISITION A search was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, on the PubMmed/MEDLINE database. The population analyzed included elderly patients undergoing pancreatic resection for pancreatic cancer as compared with younger patients to evaluate morbidity, mortality, clinically relevant postoperative pancreatic fistula, delayed gastric emptying, post-pancreatectomy hemorrhage, length of stay, overall survival, and disease-free survival. EVIDENCE SYNTHESIS The results of each study were reported using Risk Ratio (RR), Odds Ratio (OR) or Mean Difference and their P value. Twenty-four studies were included in the review for a total of 33,896 cases of which 25,937 (76.5%) were young people and 7378 (23.5%) were elderly people. The elderly age cut-off was mainly defined as ≥70 years old. Regarding the age cut-off ≥70 years old, the results comparing elderly patients (≥70 years) and younger patients (<70 years) were similar; for the age cut-offs ≥75 and ≥80 years old. Higher mortality and morbidity rates were found in the studies due to the fact that patients ≥75 and ≥80 years or older more frequently had major comorbidities than the younger patients. CONCLUSIONS This review showed that 1) elderly age cut-off has to be considered as ≥ 70 years old, and 2) age alone is not a contraindication for pancreatic cancer resection. However, elderly patients were frailer and more vulnerable than younger patients, and therefore required a careful preoperative assessment.
Collapse
Affiliation(s)
- Carlo Ingaldi
- Division of Pancreatic Surgery, IRCCS University Hospital of Bologna, Bologna, Italy
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Margherita Minghetti
- Division of Pancreatic Surgery, IRCCS University Hospital of Bologna, Bologna, Italy
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Vincenzo D'Ambra
- Division of Pancreatic Surgery, IRCCS University Hospital of Bologna, Bologna, Italy
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Claudio Ricci
- Division of Pancreatic Surgery, IRCCS University Hospital of Bologna, Bologna, Italy
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Laura Alberici
- Division of Pancreatic Surgery, IRCCS University Hospital of Bologna, Bologna, Italy
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Riccardo Casadei
- Division of Pancreatic Surgery, IRCCS University Hospital of Bologna, Bologna, Italy -
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| |
Collapse
|
91
|
Fernandes I, Macedo D, Gouveia E, Ferreira A, Lima J, Lopez D, Melo-Alvim C, Carvalho A, Tavares P, Rodrigues-Santos P, Cardoso P, Magalhães M, Vieira P, Brito J, Mendes C, Rodrigues J, Netto E, Oliveira V, Sousa C, Henriques Abreu M, Pina F, Vasques H. [Practical Guidance on the Detection of NTRK Fusions in Sarcomas: Current Status and Diagnostic Challenges]. ACTA MEDICA PORT 2025; 38:266-275. [PMID: 40185143 DOI: 10.20344/amp.21925] [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: 06/13/2024] [Accepted: 12/26/2024] [Indexed: 04/07/2025]
Abstract
Sarcomas are a rare and heterogeneous group of mesenchymal malignant tumors and account for approximately 1% of all adult cancers and around 20% of all pediatric solid tumors in Europe. Technology advances have enabled a more accurate and efficient characterization of the molecular mechanisms underlying the pathogenesis of sarcoma subtypes and revealed novel and unexpected therapeutic targets with prognostic/predictive biomarkers, namely the neurotrophic tyrosine receptor kinase (NTRK) gene fusion. The NTRK fusion assessment has recently become a standard part of management for patients with unresectable locally advanced or metastatic cancers and has been identified in various tumor types. In the more prevalent adult and pediatric sarcomas, NTRK fusions are present in 1% and 20%, respectively, and in more than 90% of very rare subsets of tumors. The inhibition of TRK activity with first-generation TRK inhibitors has been found to be effective and well tolerated in adult and pediatric patients, independently of the tumor type. Overall, the therapeutic benefit to those patients compensates for the difficulties of identifying NTRK gene fusions. However, the rarity and diagnostic complexity of NTRK gene fusions raise several questions and challenges for clinicians. To address these issues, an expert panel of medical and pediatric oncologists, radiologists, surgeons, orthopedists, and pathologists reviewed the recent literature and discussed the current status and challenges, proposing a diagnostic algorithm for identifying NTRK fusion sarcomas. The aim of this article is to review the updated information on this issue and to provide the experts' recommendations and practical guidance on the optimal management of patients with soft tissue sarcomas, infantile fibrosarcoma, gastrointestinal stromal tumors, and osteosarcoma.
Collapse
Affiliation(s)
- Isabel Fernandes
- EpiDoC Unit. Comprehensive Health Research Center (CHRC). NOVA Medical School. Universidade NOVA de Lisboa. Lisbon. Portugal
| | - Daniela Macedo
- Department of Medical Oncology. Hospital dos Lusíadas. Lisbon. Portugal
| | - Emanuel Gouveia
- Department of Medical Oncology. Instituto Português de Oncologia de Lisboa Francisco Gentil. Lisbon. Portugal
| | - Ana Ferreira
- Department of Medical Oncology. Instituto Português de Oncologia do Porto Francisco Gentil. Porto. Portugal
| | - Jorge Lima
- Instituto de Patologia e Imunologia Molecular (IPATIMUP). Universidade do Porto. Porto. Portugal
| | - Dolores Lopez
- Department of Medical Oncology. Hospital de Santa Maria. Unidade Local de Saúde Santa Maria. Lisbon. Portugal
| | - Cecília Melo-Alvim
- Department of Medical Oncology. Hospital de Santo António. Unidade Local de Saúde (ULS) de Santo António. Porto. Portugal
| | - Alice Carvalho
- Department of Pediatrics. Unidade Local de Saúde de Coimbra. Coimbra. Portugal
| | - Paulo Tavares
- Sarcoma and Bone tumors Unit. Unidade Local de Saúde de Coimbra. Coimbra. Portugal
| | - Paulo Rodrigues-Santos
- Immunology and oncology laboratory. Centro de Neurociências e Biologia Celular (CNC). Universidade de Coimbra. Coimbra. Portugal
| | - Pedro Cardoso
- Department of Orthopedics. Hospital Geral de Santo António. Unidade Local de Saúde Santo António. Porto. Portugal
| | - Manuel Magalhães
- Department of Medical Oncology. Hospital de Santo António. Unidade Local de Saúde Santo António. Porto. Portugal
| | - Paula Vieira
- Department of Medical Oncology. Hospital Dr. Nélio Mendonça. Serviço de Saúde da Região Autónoma da Madeira. Funchal. Portugal
| | - Joaquim Brito
- Department of Orthopedics. Hospital de Santa Maria. Unidade Local de Saúde Santa Maria. Lisbon. Portugal
| | - Cristina Mendes
- Department of Pediatrics. Instituto Português de Oncologia de Lisboa Francisco Gentil. Lisbon. Portugal
| | - Joana Rodrigues
- Department of Medical Oncology. Unidade Local de Saúde de Coimbra. Coimbra. Portugal
| | - Eduardo Netto
- EpiDoC Unit. Comprehensive Health Research Center (CHRC). NOVA Medical School. Universidade NOVA de Lisboa. Lisbon. Portugal; Department of Radiotherapy. Instituto Português de Oncologia de Lisboa Francisco Gentil. Lisbon. Portugal
| | - Vânia Oliveira
- Department of Orthopedics. Hospital de Santo António. Unidade Local de Saúde Santo António. Porto. Portugal
| | - Catarina Sousa
- Department of Pediatrics. Instituto Português de Oncologia do Porto Francisco Gentil. Porto. Portugal
| | - Miguel Henriques Abreu
- Department of Medical Oncology. Instituto Português de Oncologia do Porto Francisco Gentil. Porto. Portugal
| | - Filomena Pina
- Department of Radiotherapy. Hospital de Santa Maria. Unidade Local de Saúde Santa Maria. Lisbon. Portugal
| | - Hugo Vasques
- Department of General Surgery. Instituto Português de Oncologia do Porto Francisco Gentil. Porto. Portugal
| |
Collapse
|
92
|
Gu J, Liu T, Ni B, Huang Y, Shen Y, Zhang Y, Guan Y, Bai L, Zhang H, Aimaiti M, Wang S, Yue B, Xia X, Zhang Z, Cao H. A Retrospective Study of Laparoscopic Distal Gastrectomy Guided by Carbon Nanoparticle Suspension Injection Lymphography for Gastric Cancer. Am Surg 2025; 91:614-620. [PMID: 39754411 DOI: 10.1177/00031348241309565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
BackgroundThe use of lymph node (LN) tracers can help obtain a complete dissection of the LNs and increase the detection rate of metastatic LNs. Carbon nanoparticle suspension injection (CNSI) has become increasingly used in radical gastrectomy procedures. This study is designed to evaluate the quality of LN dissection in gastric cancer patients with laparoscopic distal gastrectomy under the guidance of CNSI lymphography.MethodThis was a retrospective cohort study including patients with a pathological biopsy diagnosis of resectable gastric cancer who underwent laparoscopic distal gastrectomy. Data was focused on patients at [Renji Hospital of Shanghai Jiaotong University] from July 2023 to January 2024. Patients were divided into the CNSI group and control group after 1:1 propensity score matching analysis. The median number of LNs harvested was compared between groups. Perioperative status and any complications that arose within 30 days were also analyzed.ResultAfter 1:1 propensity matching analysis, there were 49 patients each in the CNSI group and control group. The median number of harvested LNs was larger in the CNSI group than the control group (P = 0.01). A significant difference between 2 groups was observed in surgery time (P = 0.008). The morbidity of any short-term postoperative complications within 30 days after surgery revealed a similar outcome (P > 0.05).DiscussionCNSI-guided laparoscopic distal gastrectomy is less time-consuming and harvests more LNs. For laparoscopic distal gastrectomy, CNSI-guided lymphography can be an excellent adjuvant.
Collapse
Affiliation(s)
- Jiayi Gu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Liu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Ni
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yile Huang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanying Shen
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yeqian Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yujing Guan
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Long Bai
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haoyu Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Muerzhate Aimaiti
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuchang Wang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ben Yue
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiang Xia
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zizhen Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Cao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
93
|
Cheng V, Sayre EC, Cheng V, Loree JM, Gill S, Murphy RA, Howren A, De Vera MA. Mental healthcare utilisation among individuals with colorectal cancer: population-based cohort studies. BMJ ONCOLOGY 2025; 4:e000690. [PMID: 40177172 PMCID: PMC11962786 DOI: 10.1136/bmjonc-2024-000690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/10/2025] [Indexed: 04/05/2025]
Abstract
Objective Individuals with colorectal cancer (CRC) have an increased risk of mental disorders, yet mental healthcare utilisation has not been adequately examined. We evaluated mental healthcare utilisation and receipt of minimally adequate treatments for anxiety and/or depression among individuals with and without CRC. Methods and analysis We used administrative health databases from British Columbia, Canada, comprised of individuals with CRC and individuals without CRC, matched (1:1 ratio) on age, sex and incident mental disorder(s) (ie, occurring after CRC diagnosis/matched date). Primary outcomes were minimally adequate antidepressant pharmacotherapy (≥84 days' supply) and psychological (≥4 services) treatment. Results Among individuals with CRC, 1462 had incident anxiety (mean age 64.6±12.5 years, 59.2% females), 4640 had incident depression (mean age 66.3±12.3 years, 51.2% females). Approximately one in four individuals with CRC were diagnosed with anxiety (23.4%) and/or depression (23.2%) in the first year after CRC diagnosis. Minimally adequate antidepressant pharmacotherapy (36.2%) and psychological treatment (15.9%) for anxiety were significantly lower in CRC patients than in those without CRC (pharmacotherapy adjusted OR (aOR) 0.74; 95% CI 0.61, 0.88; psychological treatment aOR 0.74; 95% CI 0.58, 0.95). Similar findings were observed for depression (pharmacotherapy aOR 0.81; 95% CI 0.74, 0.90). Among individuals with CRC, mental healthcare utilisation persisted up to 10 years post-mental disorder diagnosis. Conclusions Individuals with CRC receive less mental health treatment for anxiety and/or depression, compared with those without CRC. Findings raise awareness for the need for ongoing mental healthcare throughout and beyond CRC.
Collapse
Affiliation(s)
- Vicki Cheng
- The Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
- Collaboration for Outcomes Research and Evaluation, Vancouver, British Columbia, Canada
| | - Eric C Sayre
- British Columbia Centre on Substance Use, Vancouver, British Columbia, Canada
| | - Vienna Cheng
- The Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
- Collaboration for Outcomes Research and Evaluation, Vancouver, British Columbia, Canada
| | - Jonathan M Loree
- BC Cancer, Vancouver, British Columbia, Canada
- Division of Medical Oncology, The University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada
| | - Sharlene Gill
- BC Cancer, Vancouver, British Columbia, Canada
- Division of Medical Oncology, The University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada
| | - Rachel A Murphy
- BC Cancer, Vancouver, British Columbia, Canada
- School of Population and Public Health, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Alyssa Howren
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Palo Alto, California, USA
| | - Mary A De Vera
- The Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
- Collaboration for Outcomes Research and Evaluation, Vancouver, British Columbia, Canada
- Centre for Health Evaluation and Outcome Sciences, Vancouver, British Columbia, Canada
| |
Collapse
|
94
|
Chen O, Fu L, Wang Y, Li J, Liu J, Wen Y. Targeting HSP90AA1 to overcome multiple drug resistance in breast cancer using magnetic nanoparticles loaded with salicylic acid. Int J Biol Macromol 2025; 298:139443. [PMID: 39756742 DOI: 10.1016/j.ijbiomac.2024.139443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/31/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025]
Abstract
Multiple drug resistance (MDR) remains a major obstacle in effective breast cancer chemotherapy. This study explores the role of HSP90AA1 in driving MDR and evaluates the potential of magnetic nanoparticles (Fe3O4@SA) loaded with salicylic acid (SA) to counteract drug resistance. A comprehensive screening of 200 SA-related target genes identified nine core genes, including HSP90AA1. Pharmacophore analysis revealed that SA interacts with HSP90AA1, a key regulator of mitochondrial K+ channels. Fe3O4@SA nanoparticles demonstrated efficient cellular uptake and lysosomal escape, markedly improving the chemosensitivity of resistant breast cancer cells and promoting apoptosis. In vivo experiments further confirmed the anticancer efficacy of Fe3O4@SA, highlighting its potential as a promising therapeutic strategy to overcome MDR in breast cancer.
Collapse
Affiliation(s)
- Ou Chen
- Department of clinical laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Linlin Fu
- Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yu Wang
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jinggui Li
- Liaoning Jiahe Hospital of Traditional Chinese Medicine, Medical Imaging Center, Shenyang, China
| | - Jun Liu
- Department of cardiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| | - Yanqing Wen
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
95
|
Liang P, Li Z, Chen Z, Chen Z, He F, Jin T, Cao Y, Yang K. HER2 regulates autophagy and promotes migration in gastric cancer cells through the cGAS-STING pathway. Anticancer Drugs 2025; 36:306-318. [PMID: 39869353 PMCID: PMC11884795 DOI: 10.1097/cad.0000000000001680] [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: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 01/28/2025]
Abstract
In gastric cancer, the relationship between human epidermal growth factor receptor 2 (HER2), the cyclic GMP-AMP synthase-stimulator of the interferon genes (cGAS-STING) pathway, and autophagy remains unclear. This study examines whether HER2 regulates autophagy in gastric cancer cells via the cGAS-STING signaling pathway, influencing key processes such as cell proliferation and migration. Understanding this relationship could uncover new molecular targets for diagnosis and treatment. Through lentiviral transfection, cell counting kit-8 assays, colony formation, transwell migration, scratch assays, and siRNA, we found that HER2 overexpression suppresses the cGAS-STING pathway, inhibits autophagy, and enhances the migratory ability of gastric cancer cells. In contrast, HER2 knockdown activates the cGAS-STING pathway, promotes autophagy, and reduces cell migration. We further observed that the inhibition of autophagy using chloroquine (CQ) increases the migration ability of HER2-overexpressing cells. Moreover, interfering with STING expression reversed the migration defects caused by HER2 knockdown, underscoring the critical role of the cGAS-STING pathway in HER2-regulated cell migration. We also revealed that high STING expression in gastric cancer is significantly associated with poor prognosis. STING expression was identified as an independent prognostic factor for survival (hazard ratio, 1.942; 95% confidence interval, 1.06-3.54; P = 0.031). These results highlight the importance of HER2-driven regulation of autophagy through the cGAS-STING pathway in gastric cancer progression and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Panping Liang
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zedong Li
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhengwen Chen
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zehua Chen
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Fengjun He
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Jin
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuwei Cao
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kun Yang
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
96
|
Kruiswijk AA, Vlug LAE, Acem I, Engelhardt EG, Gronchi A, Callegaro D, Haas RL, van de Wal RJP, van de Sande MAJ, van Bodegom-Vos L. Risk-Prediction Models for Clinical Decision-Making in Sarcoma Care: An International Survey Among Soft-Tissue Sarcoma Clinicians. Ann Surg Oncol 2025; 32:2958-2970. [PMID: 39893340 PMCID: PMC11882627 DOI: 10.1245/s10434-024-16849-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025]
Abstract
INTRODUCTION Risk prediction models (RPMs) are statistical tools that predict outcomes on the basis of clinical characteristics and can thereby support (shared) decision-making. With the shift toward personalized medicine, the number of RPMs has increased exponentially, including in multimodal sarcoma care. However, their integration into routine soft-tissue sarcoma (STS) care remains largely unknown. Therefore, we inventoried RPM use in sarcoma care during tumor board discussions and patient consultations as well as the attitudes toward the use of RPMs to support (shared) decision-making among STS clinicians. MATERIALS AND METHODS A 29-item survey was disseminated online to members of international sarcoma societies. RESULTS This study enrolled 278 respondents. Respectively, 68% and 65% of the clinicians reported using RPMs during tumor board discussions and/or patient consultations. During tumor board discussions, RPMs were used primarily to assess the potential benefits of (neo)adjuvant chemotherapy. During patient consultations, RPMs were used to predict patient prognosis upon request and to assist in decision-making regarding (neo)adjuvant therapies. The reliability of patient risk predicted by RPMs and the absence of guidelines regarding the use of RPMs were identified as barriers. Additionally, some clinicians questioned the applicability of estimates from RPMs to individual patients and expressed concerns about causing unnecessary anxiety when discussing prognostic outcomes. CONCLUSIONS Responding STS clinicians frequently use RPMs to support decision-making about (neo)adjuvant therapies. However, they expressed concerns about the applicability of RPM estimates to individual patients and reported challenges in communicating prognostic outcomes with patients. These findings highlight the difficulties clinicians face when integrating RPMs into patient consultations.
Collapse
Affiliation(s)
- Anouk A Kruiswijk
- Department of Biomedical Data Sciences, Medical Decision Making, Leiden University Medical Center, Leiden, The Netherlands.
- Orthopedic Surgery, Leiden University Medical Center, Leiden, The Netherlands.
| | - Lisa A E Vlug
- Department of Biomedical Data Sciences, Medical Decision Making, Leiden University Medical Center, Leiden, The Netherlands
| | - Ibtissam Acem
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Ellen G Engelhardt
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Dario Callegaro
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rick L Haas
- Department of Radiotherapy, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Radiotherapy, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Leti van Bodegom-Vos
- Department of Biomedical Data Sciences, Medical Decision Making, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
97
|
Morelli I, Greto D, Visani L, Lombardi G, Scorsetti M, Clerici E, Navarria P, Minniti G, Livi L, Desideri I. Integrating nutritional status and hematological biomarkers for enhanced prognosis prediction in glioma patients: A systematic review. Clin Nutr ESPEN 2025; 66:269-280. [PMID: 39864522 DOI: 10.1016/j.clnesp.2025.01.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
PURPOSE Multiple inflammatory and nutritional biomarkers have been established as independent prognostic factors across various solid tumors, but their role in outcomes prediction for glioma is still under investigation. Aim of the present systematic review is to report the available evidence regarding the impact of nutritional assessment and intervention for glioma prognosis and patients' quality of life (QoL). MATERIALS AND METHODS Our systematic review conformed to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. The PubMed and EMBASE databases were searched to identify studies assessing the impact of nutritional status and intervention and hematological biomarkers on survival outcomes and quality of life in patients with newly diagnosed gliomas. In the search strategy Medical Subject Headings (MeSH) terms were used. Search terms included ("nutritional status" or "nutritional assessment" or "nutritional intervention") AND ("glioma" or "glioblastoma" or "high-grade glioma" or "low-grade glioma" or "anaplastic astrocytoma" or "anaplastic oligodendroglioma") AND ("prognosis" or "survival outcomes"). The quality of each study was investigated based on the Newcastle-Ottawa Scale (NOS) criteria. Selected papers were in English and included publications in humans. This study was registered on PROSPERO (Registration No. CRD42024555442). RESULTS Our search retrieved 20 papers published between 2015 and 2023, all aiming at investigating correlations between hematological biomarkers (albumin, prealbumin, fibrinogen) and/or nutritional tools (Controlling Nutritional Score, CONUT; Prognostic Nutritional Index, PNI) and survival outcomes and quality of life of glioma patients. Nutritional intervention as well was evaluated for outcomes prediction. Overall, most papers contributed to the evidence of how nutritional assessment and inflammatory biomarkers could play an independent prognostic role also in the management of glioma patients. CONCLUSIONS PNI, CONUT score and hematological biomarkers (e.g. albumin, globulin, neutrophils, lymphocytes) may serve as useful predictors in patients with gliomas, potentially influencing clinical decisions. Additional large-scale studies are required to validate these findings and determine the mechanisms by which nutritional status, systemic inflammation and immune status affect prognosis in glioma patients.
Collapse
Affiliation(s)
- Ilaria Morelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.
| | - Daniela Greto
- Azienda Ospedaliero-Universitaria Careggi, Radiation Oncology Unit, Florence, Italy
| | - Luca Visani
- Azienda Ospedaliero-Universitaria Careggi, Radiation Oncology Unit, Florence, Italy
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Marta Scorsetti
- Radiotherapy and Radiosurgery Department, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Elena Clerici
- Radiotherapy and Radiosurgery Department, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Pierina Navarria
- Radiotherapy and Radiosurgery Department, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Giuseppe Minniti
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| | - Lorenzo Livi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Isacco Desideri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy; Azienda Ospedaliero-Universitaria Careggi, Radiation Oncology Unit, Florence, Italy
| |
Collapse
|
98
|
Park S, Haam K, Heo H, Kim D, Kim M, Jung H, Cha S, Kim M, Lee H. Integrative transcriptomic analysis identifies emetine as a promising candidate for overcoming acquired resistance to ALK inhibitors in lung cancer. Mol Oncol 2025; 19:1155-1169. [PMID: 39540457 PMCID: PMC11977641 DOI: 10.1002/1878-0261.13738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/02/2024] [Accepted: 09/10/2024] [Indexed: 11/16/2024] Open
Abstract
Anaplastic lymphoma kinase (ALK; also known as ALK tyrosine kinase receptor) inhibitors (ALKi) are effective in treating lung cancer patients with chromosomal rearrangement of ALK. However, continuous treatment with ALKis invariably leads to acquired resistance in cancer cells. In this study, we propose an efficient strategy to suppress ALKi resistance through a meta-analysis of transcriptome data from various cell models of acquired resistance to ALKis. We systematically identified gene signatures that consistently showed altered expression during the development of resistance and conducted computational drug screening using these signatures. We identified emetine as a promising candidate compound to inhibit the growth of ALKi-resistant cells. We demonstrated that emetine exhibited effectiveness in inhibiting the growth of ALKi-resistant cells, and further interpreted its impact on the resistant signatures through drug-induced RNA-sequencing data. Our transcriptome-guided systematic approach paves the way for efficient drug discovery to overcome acquired resistance to cancer therapy.
Collapse
Affiliation(s)
- Sang‐Min Park
- College of PharmacyChungnam National UniversityDaejeonKorea
| | - Keeok Haam
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
| | - Haejeong Heo
- Personalized Genomic Medicine Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
- Department of Functional GenomicsUniversity of Science and Technology (UST)DaejeonKorea
| | - Doyeong Kim
- College of PharmacyChungnam National UniversityDaejeonKorea
| | - Min‐Ju Kim
- Department of Pharmacy, College of Pharmacy and Research Institute for Drug DevelopmentPusan National UniversityBusanKorea
| | - Hyo‐Jung Jung
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
| | - Seongwon Cha
- Korean Medicine (KM) Data DivisionKorea Institute of Oriental MedicineDaejeonKorea
| | - Mirang Kim
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
- Personalized Genomic Medicine Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
- Department of Functional GenomicsUniversity of Science and Technology (UST)DaejeonKorea
| | - Haeseung Lee
- Department of Pharmacy, College of Pharmacy and Research Institute for Drug DevelopmentPusan National UniversityBusanKorea
| |
Collapse
|
99
|
Kim DH, Kim NI, Jung EK, Lee JK. Lung Adenocarcinoma Metastasis to the Vocal Cord. EAR, NOSE & THROAT JOURNAL 2025; 104:NP184-NP186. [PMID: 35722943 DOI: 10.1177/01455613221107689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Metastases to the vocal cord from a distant organ are extremely rare. This case showed metastatic adenocarcinoma on the vocal cord of lung origin in a 75-year-old nonsmoking female with a history of lung adenocarcinoma surgery 2 years earlier. The vocal cord mass was surgically removed, and the biopsy confirmed metastatic adenocarcinoma of lung origin with thyroid transcription factor (TTF)-1 positivity. A further evaluation found recurrence in the lung. The patient received gefitinib. Here, we report an extremely rare case of metastatic adenocarcinoma on the vocal cord from the lung which is the first report to our knowledge.
Collapse
Affiliation(s)
- Do Hyung Kim
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School & Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Nah Ihm Kim
- Department of Pathology, Chonnam National University Hospital, Gwangju, Korea
| | - Eun Kyung Jung
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School & Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Joon Kyoo Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School & Chonnam National University Hwasun Hospital, Hwasun, Korea
| |
Collapse
|
100
|
Guo M, Meng H, Sun Y, Zhou L, Hu T, Yu T, Bai H, Zhang Y, Gu C, Yang Y. Bruceine A Inhibits Cell Proliferation by Targeting the USP13/PARP1 Signalling Pathway in Multiple Myeloma. Basic Clin Pharmacol Toxicol 2025; 136:e70027. [PMID: 40151951 PMCID: PMC11955937 DOI: 10.1111/bcpt.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025]
Abstract
Multiple myeloma (MM) is an incurable hematologic malignancy, driving significant interest in the discovery of novel therapeutic strategies. Bruceine A (BA), a tetracyclic triterpene quassinoid derived from Brucea javanica, has shown anticancer properties by modulating multiple intracellular signalling pathways and exhibiting various biological effects. However, the specific pharmacological mechanisms by which it combats MM remain unclear. In this study, we identified USP13 as a potential target of BA. We observed a significant increase in USP13 expression in patients with MM, which was strongly associated with a poorer prognosis. Furthermore, enhanced USP13 expression can stimulate MM cell proliferation both in vitro and in vivo. Mass spectrometry analysis, combined with co-immunoprecipitation and in vitro ubiquitination experiments, revealed PARP1 as a critical downstream target of USP13. USP13 can stabilize PARP1 protein through deubiquitination, promoting PARP1-mediated DNA damage repair (DDR) and facilitating MM progression. Notably, we utilized MM cell lines, an MM Patient-Derived Tumour Xenograft model, and a 5TMM3VT mouse model to determine the anticancer effects of BA on MM progression, revealing its potential to target USP13/PARP1 signalling and disrupt DDR in MM cells. In conclusion, these findings suggest that BA inhibiting USP13/PARP1-mediated DDR might be a promising therapeutic strategy for MM.
Collapse
Affiliation(s)
- Mengjie Guo
- Nanjing Hospital of Chinese Medicine Affiliated With Nanjing University of Chinese MedicineNanjingChina
- School of MedicineNanjing University of Chinese MedicineNanjingChina
| | - Han Meng
- School of MedicineNanjing University of Chinese MedicineNanjingChina
| | - Yi Sun
- School of MedicineNanjing University of Chinese MedicineNanjingChina
| | - Lianxin Zhou
- School of MedicineNanjing University of Chinese MedicineNanjingChina
| | - Tingting Hu
- School of MedicineNanjing University of Chinese MedicineNanjingChina
| | - Tianyi Yu
- School of MedicineNanjing University of Chinese MedicineNanjingChina
| | - Haowen Bai
- School of MedicineNanjing University of Chinese MedicineNanjingChina
| | - Yuanjiao Zhang
- School of MedicineNanjing University of Chinese MedicineNanjingChina
| | - Chunyan Gu
- Nanjing Hospital of Chinese Medicine Affiliated With Nanjing University of Chinese MedicineNanjingChina
- School of MedicineNanjing University of Chinese MedicineNanjingChina
| | - Ye Yang
- School of MedicineNanjing University of Chinese MedicineNanjingChina
| |
Collapse
|