251
|
Li M, Zhang X, Zhou C, Lin L, Miao L, Wu H, He J. Neuroblastoma risk decreased by NSUN3 rs7653521 C>T polymorphism in Chinese children. Chin Med J (Engl) 2025:00029330-990000000-01449. [PMID: 40008824 DOI: 10.1097/cm9.0000000000003512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Indexed: 02/27/2025] Open
Affiliation(s)
- Meng Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong 510623, China
| | - Xinxin Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong 510623, China
| | - Chunlei Zhou
- Department of Pathology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Lei Lin
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong 510623, China
| | - Lei Miao
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong 510623, China
| | - Haiyan Wu
- Department of Pathology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong 510623, China
| |
Collapse
|
252
|
Ghimire K, Awasthi BP, Yadav K, Lee J, Kim H, Jeong BS, Kim JA. Prostate cancer-selective anticancer action of an oxindole derivative via HO-1-mediated disruption of metabolic reprogramming. Chem Biol Interact 2025; 408:111393. [PMID: 39842705 DOI: 10.1016/j.cbi.2025.111393] [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: 11/13/2024] [Revised: 12/26/2024] [Accepted: 01/20/2025] [Indexed: 01/24/2025]
Abstract
Prostate cancer, the second leading cause of cancer-related mortality in men, exhibits distinct metabolic reprogramming involving zinc and citrate metabolism. This study investigated whether targeting this unique metabolic profile could offer an effective therapeutic approach. A series of novel oxindole derivatives were synthesized and evaluated for their inhibitory effects on transcription factors (TFs) and antiproliferative activity across various cancer cell lines. Among these, compound 3D showed the strongest inhibition of master TFs (HIF-1α, c-Myc, and SP-1) and demonstrated selective antiproliferative activity in prostate cancer cells. In PC-3 and LNCaP cells, compound 3D suppressed aerobic glycolysis by downregulating lactate-modulating genes (LDHA, MCT1/4, and CAIX) and the zinc influx transporter (ZIP1), without affecting the zinc efflux transporter (ZnT4). Notably, 3D selectively increased heme oxygenase-1 (HO-1) levels in prostate cancer cells, as shown by the proteome profiler oncogene array assay and confirmed by Western blotting. This response was reversed by ZnCl2 treatment. The decreases in LDHA, mitochondrial mass (measured by FACS), and cell proliferation induced by compound 3D were blocked by HO-1-IN-1, an HO-1 inhibitor, and ZnCl2. Furthermore, 3D induced a more pronounced reduction in the oxygen consumption rate (OCR) than in the extracellular acidification rate (EACR), indicating a strong effect on oxidative metabolism. 3D exhibited dose-dependent antitumour efficacy in vivo comparable to that of docetaxel. These findings reveal that the oxindole derivative 3D substantially lowers intracellular zinc levels, yielding potent antitumour effects in prostate cancer through HO-1 upregulation, which impairs mitochondrial function more significantly than aerobic glycolysis.
Collapse
Affiliation(s)
- Kalpana Ghimire
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | | | - Kiran Yadav
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Jiwoo Lee
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Hyunjin Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Byeong-Seon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea.
| | - Jung-Ae Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea.
| |
Collapse
|
253
|
Glueck V, Grimm C, Postl M, Brueffer C, Segui N, Alcaide M, Oton L, Chen Y, Saal LH, Hofstetter G, Polterauer S, Muellauer L. ctDNA as an Objective Marker for Postoperative Residual Disease in Primary Advanced High-Grade Serous Ovarian Cancer. Cancers (Basel) 2025; 17:786. [PMID: 40075633 PMCID: PMC11899276 DOI: 10.3390/cancers17050786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/11/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES The surgeon's subjective intraoperative evaluation is the standard of care to assess postoperative residual disease (RD) in advanced epithelial ovarian cancer (EOC). We investigated the feasibility of ctDNA as an objective marker for postoperative RD. METHODS This prospective study included 27 patients with advanced ovarian cancer (FIGO IIIA1-IVB) who underwent primary surgery between July 2021 and July 2022. Blood samples were analyzed preoperatively and on days 2 (d2) and 10 (d10) postoperatively. Low-coverage whole genome sequencing (WGS) was used to identify structural variants (SVs) at single-base pair resolution, single nucleotide variants (SNVs), and indels in tumor tissue to develop personalized, tumor-informed digital polymerase chain reaction (dPCR) fingerprint assays for each patient. RESULTS dPCR fingerprint assays were successfully developed for all patients by identifying one to eight SVs/SNVs per patient. ctDNA was detected in 96% (n = 26/27) of patients preoperatively and in 81% (n = 22/27) of patients at d10. Median ctDNA levels at d10 were significantly higher in patients with postoperative RD (median 367.38 copies (cps)/mL, 2.84% variant allele frequency; VAF) than in patients without postoperative RD (median 0.92 cps/mL, 0.017% VAF, p < 0.001). In patients with postoperative RD, ctDNA levels increased from the preoperative stage to d10 in seven out of eight patients (p = 0.016). In patients with complete tumor resection, ctDNA levels decreased from the preoperative stage to d10 in 17/19 patients (p < 0.001). CONCLUSIONS A tumor-informed personalized ctDNA approach demonstrated feasibility, providing extremely high detection rates pre- and postoperatively. These results indicate that this approach could potentially be used for postoperative RD assessment in patients with primary advanced EOC.
Collapse
Affiliation(s)
- Valentina Glueck
- Gynecologic Cancer Unit, Division of General Gynecology and Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.); (S.P.)
- Department of Obstetrics and Gynecology, Klinikum Starnberg, 82319 Starnberg, Germany
| | - Christoph Grimm
- Gynecologic Cancer Unit, Division of General Gynecology and Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.); (S.P.)
| | - Magdalena Postl
- Gynecologic Cancer Unit, Division of General Gynecology and Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.); (S.P.)
| | - Christian Brueffer
- SAGA Diagnostics AB, 223 81 Lund, Sweden; (C.B.); (N.S.); (M.A.); (L.O.); (Y.C.); (L.H.S.)
- Division of Oncology, Lund University Cancer Center, Skåne University Hospital Comprehensive Cancer Center, Lund University, 221 00 Lund, Sweden
| | - Nuria Segui
- SAGA Diagnostics AB, 223 81 Lund, Sweden; (C.B.); (N.S.); (M.A.); (L.O.); (Y.C.); (L.H.S.)
| | - Miguel Alcaide
- SAGA Diagnostics AB, 223 81 Lund, Sweden; (C.B.); (N.S.); (M.A.); (L.O.); (Y.C.); (L.H.S.)
| | - Lucia Oton
- SAGA Diagnostics AB, 223 81 Lund, Sweden; (C.B.); (N.S.); (M.A.); (L.O.); (Y.C.); (L.H.S.)
| | - Yilun Chen
- SAGA Diagnostics AB, 223 81 Lund, Sweden; (C.B.); (N.S.); (M.A.); (L.O.); (Y.C.); (L.H.S.)
| | - Lao H. Saal
- SAGA Diagnostics AB, 223 81 Lund, Sweden; (C.B.); (N.S.); (M.A.); (L.O.); (Y.C.); (L.H.S.)
- Division of Oncology, Lund University Cancer Center, Skåne University Hospital Comprehensive Cancer Center, Lund University, 221 00 Lund, Sweden
| | - Gerda Hofstetter
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (G.H.); (L.M.)
| | - Stephan Polterauer
- Gynecologic Cancer Unit, Division of General Gynecology and Gynecologic Oncology, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.); (S.P.)
| | - Leonhard Muellauer
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (G.H.); (L.M.)
| |
Collapse
|
254
|
Uysal M, Wehrle CJ, Coppa C, Kamath S, Krishnamurthi S, Martin C, Hag ME, Khalil M, Fujiki M, Schlegel A, Miller C, Hashimoto K, Aucejo F, Kwon DC, Kim J. Bridging therapy with histotripsy prior to liver transplantation for hepatocellular carcinoma: a first case report. Exp Hematol Oncol 2025; 14:20. [PMID: 40001149 PMCID: PMC11863397 DOI: 10.1186/s40164-025-00604-z] [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: 11/12/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Histotripsy is a novel, non-invasive, non-ionizing, and non-thermal ablation technique that disrupts tumors using acoustic cavitation. We report the first use of Histotripsy as bridging therapy prior to liver transplant for hepatocellular carcinoma (HCC) treated with histotripsy. CASE PRESENTATION A 59-year-old woman presented with Metabolic-Associated Steatotic Liver Disease (MASLD) cirrhosis (labMELD = 14), hepatic encephalopathy, and a single 2 cm OPTN V lesion in the left lateral segment consistent with HCC. The patient underwent histotripsy treatment of the lesion as bridging therapy before receiving liver transplantation. Histopathology analysis of the explanted liver showed total necrosis of the treated area, with no residual viable tissue tumor. CONCLUSION This case demonstrates the potential utility of histotripsy as an effective bridging therapy for patients with combined cirrhosis and hepatocellular carcinoma (HCC) awaiting liver transplantation, with complete tumor necrosis on explant pathology demonstrating its therapeutic efficacy.
Collapse
Affiliation(s)
- Melis Uysal
- Department of General Surgery, Digestive Disease & Surgery Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Chase J Wehrle
- Department of General Surgery, Digestive Disease & Surgery Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | | | - Suneel Kamath
- Department of Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH, USA
| | - Smitha Krishnamurthi
- Department of Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH, USA
| | | | - Mohamed El Hag
- Diagnostics Institute, Department of Pathology, Cleveland Clinic, Cleveland, OH, USA
| | - Mazhar Khalil
- Department of General Surgery, Digestive Disease & Surgery Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Masato Fujiki
- Department of General Surgery, Digestive Disease & Surgery Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Andrea Schlegel
- Department of General Surgery, Digestive Disease & Surgery Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
- Department of Inflammation and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, USA
| | - Charles Miller
- Department of General Surgery, Digestive Disease & Surgery Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Koji Hashimoto
- Department of General Surgery, Digestive Disease & Surgery Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Federico Aucejo
- Department of General Surgery, Digestive Disease & Surgery Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - David Ch Kwon
- Department of General Surgery, Digestive Disease & Surgery Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Jaekeun Kim
- Department of General Surgery, Digestive Disease & Surgery Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
| |
Collapse
|
255
|
Wang J, Cai J, Wang Z, Yang S, Wang J, Jia Y, Sun H, Ma X. α5-nAChR/NETO2 contributed to chronic stress-promoted lung adenocarcinoma progression. Cancer Cell Int 2025; 25:67. [PMID: 40001189 PMCID: PMC11853797 DOI: 10.1186/s12935-025-03701-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND α5-nicotinic acetylcholine receptor (α5-nAChR) participates in chronic stress-promoted lung adenocarcinoma (LUAD) progression. Neuropilin and tolloid-like 2 (NETO2) contributes to fear expression and extinction, which is related to tumorigenesis. CHRNA5 (encoding α5-nAChR) gene profiling revealed a reduction in NETO2 expression following CHRNA5 knockdown. Nevertheless, the connection between α5-nAChR and NETO2 in LUAD progression induced by chronic stress remains unclear. METHODS RNA-Seq and bioinformatics database were used for analyzing the expression as well as correlation of α5-nAChR, together with NETO2 in LUAD. α5-nAChR and NETO2 expression were detected using immunohistochemistry in LUAD tissue microarrays, chronic restraint stress (CRS) and chronic unpredictable stress (CUMS) mice tissues. In lung adenocarcinoma A549 and H1299 cells, the expression of α5-nAChR, NETO2, p-CAMKII, p-STAT3 and vimentin induced by acetylcholine/nicotine was examined by western blot. The interaction of α5-nAChR with NETO2 in lung adenocarcinoma cells was detected by Co-immunoprecipitation assay and modeled using molecular docking. EdU assay and colony formation assay were conducted to evaluate cell proliferation, while wound healing assay as well as transwell assay assessed the migration and invasion of lung adenocarcinoma cells. RESULTS α5-nAChR expression was related to NETO2 expression, low survival rate, staging as well as smoking status in LUAD dataset as well as tissue microarrays. The correlation between α5-nAChR and NETO2 was validated in nude mice xenograft tissues. α5-nAChR as well as NETO2 expression correlated in CRS and CUMS mice tissues. In vitro, acetylcholine/nicotine mediated NETO2, p-CAMKII, p-STAT3 and vimentin expression via α5-nAChR. α5-nAChR interacted with NETO2 as well as CAMKII in LUAD cells. α5-nAChR/NETO2 signaling contributed to LUAD cell proliferation, migration and invasion. CONCLUSIONS The above results uncover a new chronic stress-promoted LUAD signaling pathway: α5-nAChR/NETO2 axis contributes to chronic stress-promoted LUAD cell proliferation, migration and invasion.
Collapse
Affiliation(s)
- Jingting Wang
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013, China
| | - Jiaying Cai
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013, China
| | - Zengping Wang
- Department of Medical Laboratory, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Shuran Yang
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013, China
| | - Jing Wang
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013, China
| | - Yanfei Jia
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013, China
| | - Haiji Sun
- College of Life Science, Shandong Normal University, Shandong, 250014, China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013, China.
- Department of Medical Laboratory, Shandong Second Medical University, Weifang, Shandong, 261053, China.
| |
Collapse
|
256
|
Komatsu S, Nagamata S, Terashima K, Demizu Y, Suga M, Kido M, Yanagimoto H, Toyama H, Tokumaru S, Okimoto T, Terai Y, Fukumoto T. Combination Treatment with Spacer Placement Surgery Followed by Particle Radiotherapy for Lymph Node Metastasis from Uterine Cancer. Ann Surg Oncol 2025:10.1245/s10434-025-17039-9. [PMID: 40000562 DOI: 10.1245/s10434-025-17039-9] [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: 11/12/2024] [Accepted: 02/04/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND The effectiveness of local treatment in lymph node metastasis from uterine cancer has been proven; the standard treatment is surgical intervention. Although radiotherapy, including particle radiotherapy (PRT), is an alternative local treatment, its application is often contraindicated owing to its proximity to the gastrointestinal tract. Combination treatment with spacer placement surgery followed by PRT is a potential solution to this problem. This study aimed to evaluate the outcomes of this combination treatment of lymph node metastases from uterine cancer. PATIENTS AND METHODS Between December 2007 and March 2023, ten consecutive patients who underwent combination treatment comprising spacer placement surgery and subsequent PRT were assessed for treatment outcomes. RESULTS The median survival time was 53.5 months; the 3- and 5-year overall survival rates were 76.2% and 38.1%, respectively. The 3- and 5-year local control rates in all patients were both 88.9%. The median volume irradiated at 95% of the treatment planning dose (V95%) of the gross tumor volume, clinical target volume, and planning target volume were 100.0%, 99.8%, and 92.2%, respectively. The median dose intensity covering 95% of the target volume (D95%) of the gross tumor volume/planned dose, clinical target volume/planned dose, and planning target volume/planned dose were 98.9%, 99.0%, and 87.2%, respectively. CONCLUSIONS Spacer placement surgery contributed to the optimized PRT dose distribution and might have contributed to favorable local control and survival rates. This innovative combination treatment might have a significant effect on the treatment of lymph node metastases from uterine cancers.
Collapse
Affiliation(s)
- Shohei Komatsu
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Satoshi Nagamata
- Department of Obstetrics and Gynecology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazuki Terashima
- Department of Radiology, Hyogo Ion Beam Medical Center, Tatsuno, Japan
| | - Yusuke Demizu
- Department of Radiology, Hyogo Ion Beam Medical Center, Tatsuno, Japan
- Department of Radiation Oncology, Hyogo Ion Beam Medical Center Kobe Proton Center, Kobe, Japan
| | - Masaki Suga
- Department of Radiation Physics, Hyogo Ion Beam Medical Center, Tatsuno, Japan
| | - Masahiro Kido
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroaki Yanagimoto
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hirochika Toyama
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Sunao Tokumaru
- Department of Radiology, Hyogo Ion Beam Medical Center, Tatsuno, Japan
| | - Tomoaki Okimoto
- Department of Radiology, Hyogo Ion Beam Medical Center, Tatsuno, Japan
| | - Yoshito Terai
- Department of Obstetrics and Gynecology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takumi Fukumoto
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
257
|
Hu ZY, Ding D, Song Y, Deng YF, Zhang CM, Yu T. Molecular mechanism of pancreatic ductal adenocarcinoma: The heterogeneity of cancer-associated fibroblasts and key signaling pathways. World J Clin Oncol 2025; 16:97007. [PMID: 39995552 PMCID: PMC11686552 DOI: 10.5306/wjco.v16.i2.97007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/04/2024] [Accepted: 11/04/2024] [Indexed: 12/11/2024] Open
Abstract
Pancreatic ductal adenocarcinoma stands out as an exceptionally fatal cancer owing to the complexities associated with its treatment and diagnosis, leading to a notably low five-year survival rate. This study offers a detailed exploration of epidemiological trends in pancreatic cancer and key molecular drivers, such as mutations in CDKN2A, KRAS, SMAD4, and TP53, along with the influence of cancer-associated fibroblasts (CAFs) on disease progression. In particular, we focused on the pivotal roles of signaling pathways such as the transforming growth factor-β and Wnt/β-catenin pathways in the development of pancreatic cancer and investigated their application in emerging therapeutic strategies. This study provides new scientific perspectives on pancreatic cancer treatment, especially in the development of precision medicine and targeted therapeutic strategies, and demonstrates the importance of signaling pathway research in the development of effective therapeutic regimens. Future studies should explore the subtypes of CAFs and their specific roles in the tumor microenvironment to devise more effective therapeutic methods.
Collapse
Affiliation(s)
- Zhong-Yuan Hu
- First School of Clinical Medicine, Shaanxi University of Chinese Medicine, Xianyang 712000, Shaanxi Province, China
| | - Ding Ding
- First School of Clinical Medicine, Shaanxi University of Chinese Medicine, Xianyang 712000, Shaanxi Province, China
| | - Yu Song
- College of Acupuncture and Massage, Shaanxi University of Chinese Medicine, Xianyang 712000, Shaanxi Province, China
| | - Ya-Feng Deng
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou 510000, Guangdong Province, China
| | - Cheng-Ming Zhang
- Digestive Department I, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi’an 710000, Shaanxi Province, China
| | - Tao Yu
- Digestive Department I, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi’an 710000, Shaanxi Province, China
| |
Collapse
|
258
|
Feng JN, Chen PR, Yuan SF, Dai Q, Zheng WE, Lin HL. Pingxiao pian attenuate invasiveness and proliferation of lung adenocarcinoma through regulating miR-29b-3p/TGF-β1/Smad/EMT pathway. Discov Oncol 2025; 16:232. [PMID: 39992570 PMCID: PMC11850683 DOI: 10.1007/s12672-025-01959-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
INTRODUCTION Lung cancer is a highly prevalent and deadly disease worldwide, causing over 1.2 million deaths each year. Pingxiao Pian (PXP) tablets, a Chinese traditional medicine, have been widely applied in the treatment of lung cancer. However, the mechanism underlying therapeutic effects of PXP tablets remains undisclosed. METHODS A549 human LUAD cell line was utilized for in vitro experiments. Transfection of miR-29b mimic was performed using Lipofectamine 3000. PXP was purchased and dissolved into PBS and drinking water after carefully removing the outer coating. Dual-luciferase reporter assay was conducted to assess the regulatory effect of miR-29b on TGF-β1. The protein levels of epithelial-mesenchymal transition (EMT) markers and activation of TGF-β1 pathway were characterized using immunoblotting analysis. RESULTS PXP reduced the invasiveness and proliferation of LUAD cells by increasing miR-29b-3p expression in vitro. Overexpression of miR-29b-3p resulted in decreased cell proliferation and invasiveness, while silencing of miR-29b-3p in the A549 cells displayed the opposite effect. Moreover, PXP treatment reversed the increased cell proliferating rate triggered by miR-29b-3p silencing. Additionally, PXP was found to hamper EMT occurrence in A549 cells by regulating miR-29b-3p and reduce expression of N-cad and vimentin. Overexpression of miR-29b-3p blocked the phosphorylation of Smad2/3 and decreased TGF-β1 expression. Luciferase assay results indicated that miR-29b-3p directly regulated TGF-β1 expression. In vivo tumor formation experiments confirmed the tumor-reducing effects of PXP and the role of miR-29b in tumor progression. PXP treatment decreased tumor size and weight via regulating miR-29b-3p. CONCLUSION Our study suggests that PXP exerts anti-tumor effects in LUAD through the regulation of miR-29b and the inhibition of EMT via the TGF-β1/Smad2/3 pathway.
Collapse
Affiliation(s)
- Jie-Ni Feng
- Department of Medical Oncology, Rui'an People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Pei-Rui Chen
- Department of Medical Oncology, Rui'an People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Shao-Fei Yuan
- Department of Medical Oncology, Rui'an People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Qiang Dai
- Department of Medical Oncology, Rui'an People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Wei-E Zheng
- Department of Chemoradiation and Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Hua-Long Lin
- Department of Medical Oncology, Rui'an People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China.
| |
Collapse
|
259
|
Kalender E, Ekinci E, Elboğa U, Şahin E. Efficacy of 177Lu-PSMA-617 Therapy in mCRPC Patients with Liver Metastases: Insights into Survival Outcomes and Predictors of Response. Biomedicines 2025; 13:569. [PMID: 40149546 PMCID: PMC11939846 DOI: 10.3390/biomedicines13030569] [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: 01/30/2025] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025] Open
Abstract
Objectives: Metastatic castration-resistant prostate cancer (mCRPC) is associated with poor prognosis, particularly in cases of liver metastases. 177Lu-PSMA-617 (commercially known as Pluvicto) is an FDA-approved radioligand therapy for mCRPC patients. This study aimed to evaluate the efficacy of 177Lu-PSMA-617 radioligand therapy (RLT) in mCRPC patients with liver metastases, focusing on progression-free survival (PFS), overall survival (OS), and factors influencing treatment response. Materials and Methods: This retrospective study included mCRPC patients (n = 32) with liver metastases treated with Lu-PSMA-617. Patient data, including prostate-specific antigen (PSA) levels, liver SUVmax values, Lutetium-PSMA therapy cycles, and survival outcomes, were collected. Kaplan-Meier survival analysis was used to calculate PFS and OS, while regression analysis was employed to identify factors associated with treatment response. Results: The median PFS and OS were 6 and 9 months, respectively. Partial regression was observed in patients with significantly lower PSA levels (median: 90.0 ng/mL, range: 22-699 ng/mL, p = 0.001) and liver SUVmax values (median: 17.9, range: 8.3-57.0, p = 0.008). A higher number of Lutetium-PSMA cycles correlated with improved treatment response (p = 0.010) and reduced liver SUVmax values (p = 0.043). Conclusions: Lu-PSMA-617 therapy is effective in managing mCRPC with liver metastases. Increased intensity of therapy exposure, reflected by a higher number of treatment cycles, is associated with a greater biochemical response, as indicated by reduced PSA levels, thereby supporting the rationale for personalized treatment strategies. These findings support the use of Lu-PSMA-617 in mCRPC patients with liver metastases, warranting further prospective studies.
Collapse
Affiliation(s)
- Ebuzer Kalender
- Department of Nuclear Medicine, School of Medicine, Gaziantep University, Gaziantep 27310, Turkey; (E.E.); (U.E.); (E.Ş.)
| | | | | | | |
Collapse
|
260
|
Cui Y, Sun Y, Liang N, Tian C. YY1-mediated DUXAP8 facilitates HCC progression via modulating DEPDC1 expression. Clin Exp Med 2025; 25:65. [PMID: 39992478 PMCID: PMC11850567 DOI: 10.1007/s10238-025-01572-8] [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: 12/16/2024] [Accepted: 01/17/2025] [Indexed: 02/25/2025]
Abstract
The long noncoding RNA DUXAP8 has been implicated in the progression of various malignancies, including hepatocellular carcinoma (HCC). Although there is increasing evidence of DUXAP8's role in tumor biology, the exact mechanisms by which it affects the development and treatment of HCC are still unclear. Previous studies have suggested a potential link between DUXAP8 expression and disease progression, necessitating further investigation into its roles and underlying mechanisms. To clarify how DUXAP8 is involved in HCC, we measured its expression in HCC cell lines and tissues from patients. We utilized in vitro assays to evaluate the effects of DUXAP8 on tumor cell proliferation and metastasis. Additionally, we examined the regulatory relationships between DUXAP8, YY1, and DEPDC1 using RNA immunoprecipitation and luciferase reporter assays to investigate their functional mechanisms. Our findings demonstrated that DUXAP8 is frequently upregulated in HCC specimens and that its overexpression significantly enhances both the proliferation and metastatic capability of HCC cells. Importantly, the expression levels of DUXAP8, YY1, and DEPDC1 showed correlations with clinical parameters such as disease stage and histopathological characteristics. Mechanistically, we uncovered that YY1 regulates DUXAP8, which, in turn, modulates DEPDC1 expression through a dual mechanism involving the sponging of miR-7-5p and the stabilization of DEPDC1 mRNA facilitated by HNRNPF. Our study identifies DUXAP8 as a pivotal factor in the proliferation and metastasis of HCC, acting through the DUXAP8/miR-7-5p and DUXAP8/HNRNPF pathways to regulate DEPDC1 expression. These findings indicate that targeting DUXAP8 could be a new therapeutic strategy for treating HCC. Further research in both preclinical and clinical settings is needed to evaluate its potential as a biomarker and therapeutic target in liver cancer management.
Collapse
Affiliation(s)
- Yi Cui
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Yong Sun
- Department of Hepatobiliary Gastrointestinal Surgery, People's Hospital of YanHe, Tongren, China
| | - Na Liang
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Chuan Tian
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China.
| |
Collapse
|
261
|
Huan Y, She P, Cai X, Qi J, Zhang C. Identification of key biomarkers in breast cancer based on bioinformatics analysis and experimental verification. J Egypt Natl Canc Inst 2025; 37:5. [PMID: 39988630 DOI: 10.1186/s43046-025-00260-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 02/01/2025] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND Breast cancer (BC) is a malignant tumor characterized by a high incidence rate and is the leading cause of cancer-related deaths among women worldwide. This study aims to identify key genes and potential prognostic biomarkers using a bioinformatics approach. METHODS Three microarray datasets, GSE86374, GSE120129, and GSE29044, were downloaded from the GEO database. GEO2R and Venn diagram software were employed to identify differentially expressed genes (DEGs), while DAVID was utilized for functional enrichment analysis. Subsequently, STRING and Cytoscape were used to construct the protein-protein interaction (PPI) network among the DEGs. UALCAN, GEPIA, and the Kaplan-Meier plotter were employed for prognostic analysis. Following this, the correlations and alterations of key genes were examined using cBioPortal. Finally, immunohistochemistry (IHC) was performed to validate the expression levels of the key genes. RESULTS A total of 323 differentially expressed genes (DEGs) were identified. From the protein-protein interaction (PPI) network, 37 hub genes were selected. Validation using UALCAN, GEPIA, and Kaplan-Meier plotters revealed that three key genes-RACGAP1, SPAG5, and KIF20A-were significantly overexpressed and associated with poor prognosis in breast cancer (BC), as well as advanced tumor staging. The correlations and alterations of these key genes, as demonstrated on cBioPortal, indicated that their alterations co-occurred. Experimental verification through immunohistochemistry (IHC) confirmed that the proteins of these key genes were highly expressed in tumor tissues. CONCLUSIONS The key genes identified in this study can enhance our understanding of the molecular mechanisms underlying breast cancer (BC). Additionally, these genes may serve as potential sensitive biomarkers for patients with BC.
Collapse
Affiliation(s)
- Yu Huan
- Department of Clinical Laboratory, Jiading District Maternal and Children Health Hospital, Shanghai, 201821, People's Republic of China
| | - Ping She
- School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, Shanxi, 710049, People's Republic of China
| | - Xushan Cai
- Department of Clinical Laboratory, Jiading District Maternal and Children Health Hospital, Shanghai, 201821, People's Republic of China.
| | - Jiehua Qi
- Department of Clinical Laboratory, Jiading District Maternal and Children Health Hospital, Shanghai, 201821, People's Republic of China
| | - Chunli Zhang
- Department of Clinical Laboratory, Jiading District Maternal and Children Health Hospital, Shanghai, 201821, People's Republic of China
| |
Collapse
|
262
|
Chen X, Sun F, Wang X, Feng X, Aref AR, Tian Y, Ashrafizadeh M, Wu D. Inflammation, microbiota, and pancreatic cancer. Cancer Cell Int 2025; 25:62. [PMID: 39987122 PMCID: PMC11847367 DOI: 10.1186/s12935-025-03673-6] [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: 10/14/2024] [Accepted: 02/04/2025] [Indexed: 02/24/2025] Open
Abstract
Pancreatic cancer (PC) is a malignancy of gastrointestinal tract threatening the life of people around the world. In spite of the advances in the treatment of PC, the overall survival of this disease in advanced stage is less than 12%. Moreover, PC cells have aggressive behaviour in proliferation and metastasis as well as capable of developing therapy resistance. Therefore, highlighting the underlying molecular mechanisms in PC pathogenesis can provide new insights for its treatment. In the present review, inflammation and related pathways as well as role of gut microbiome in the regulation of PC pathogenesis are highlighted. The various kinds of interleukins and chemokines are able to regulate angiogenesis, metastasis, proliferation, inflammation and therapy resistance in PC cells. Furthermore, a number of molecular pathways including NF-κB, TLRs and TGF-β demonstrate dysregulation in PC aggravating inflammation and tumorigenesis. Therapeutic regulation of these pathways can reverse inflammation and progression of PC. Both chronic and acute pancreatitis have been shown to be risk factors in the development of PC, further highlighting the role of inflammation. Finally, the composition of gut microbiota can be a risk factor for PC development through affecting pathways such as NF-κB to mediate inflammation.
Collapse
Affiliation(s)
- XiaoLiang Chen
- Department of General Surgery and Integrated Traditional Chinese and Western Medicine Oncology, Tiantai People'S Hospital of Zhejiang Province(Tiantai Branch of Zhejiang Provincial People'S Hospital), Hangzhou Medical College, Taizhou, Zhejiang, China
| | - Feixia Sun
- Nursing Department, Shandong First Medical University Affiliated Occupational Disease Hospital (Shandong Provincial Occupational Disease Hospital), Jinan, China
| | - Xuqin Wang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401120, China
| | - Xiaoqiang Feng
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, 525200, Guangdong, China
| | - Amir Reza Aref
- VitroVision Department, DeepkinetiX, Inc, Boston, MA, USA
| | - Yu Tian
- Research Center, the Huizhou Central People'S Hospital, Guangdong Medical University, Huizhou, Guangdong, China.
- School of Public Health, Benedictine University, No. 5700 College Road, Lisle, IL, 60532, USA.
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China.
| | - Dengfeng Wu
- Department of Emergency, The People'S Hospital of Gaozhou, No. 89 Xiguan Road, Gaozhou, 525200, Guangdong, China.
| |
Collapse
|
263
|
Zhang Y, Li H, Zhao Y, Liu L, Zhou Y, Pan X, Ding Y, Liao W, Qi L, Huang C, Tang N. Macrocarpal I induces immunogenic cell death and synergizes with immune checkpoint inhibition by targeting tubulin and PARP1 in colorectal cancer. Cell Death Discov 2025; 11:73. [PMID: 39987121 PMCID: PMC11846858 DOI: 10.1038/s41420-025-02360-9] [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: 06/19/2024] [Revised: 01/26/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025] Open
Abstract
Colorectal cancer (CRC) presents an obstacle to immunotherapy, primarily because most cases are microsatellite stable (MSS) tumors, which are often described as "cold tumors" with limited immunogenicity. Recent studies have indicated that several therapeutic approaches, such as chemotherapy and targeted therapies, can elicit immunogenic cell death (ICD) and stimulate immune responses. However, challenges such as target affinity and in vivo pharmacokinetics limit the efficacy and immune response of current targeted therapies. In this study, we demonstrate that Macrocarpal I is a potent inducer of ICD by activating the PERK/eIF2A/ATF4/CHOP signaling pathway. Furthermore, Macrocarpal I induces apoptosis and ferroptosis, both of which act as triggers for ICD. Mechanistically, Macrocarpal I directly targets TUBB2B and PARP1, disrupting microtubule polymerization and DNA repair processes. Importantly, treatment with Macrocarpal I enhances the anti-tumor immune response and augments responsiveness to anti-PD-1 therapy in an MC38 syngeneic mouse model of CRC.
Collapse
Affiliation(s)
- Yaxin Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Huali Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yali Zhao
- Department of Pathology, Shenzhen Longgang Central Hospital, Shenzhen, 518100, China
| | - Lingtao Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yi Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xingyan Pan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yanqing Ding
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wenting Liao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Lu Qi
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Chengmei Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Na Tang
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, 518020, China.
| |
Collapse
|
264
|
Öz H, Canacankatan N, Antmen ŞE, Aytan H, Tuncel F. 'Investigation of miRNAs That Affect the PI3K/AKT/mTOR Signaling Pathway in Endometrial Cancer'. Cell Biochem Biophys 2025:10.1007/s12013-025-01694-6. [PMID: 39982560 DOI: 10.1007/s12013-025-01694-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2025] [Indexed: 02/22/2025]
Abstract
Endometrial cancer is a prevalent type of cancer among women worldwide. The irregularity of the PI3K/AKT/mTOR signaling pathway plays a role in the pathogenesis of many cancer types. MicroRNAs are small noncoding RNAs that play crucial roles in the pathogenesis of different cancer types. MicroRNAs target many key components of the PI3K/AKT/mTOR pathway in human tumors. In this study the PI3K/AKT/mTOR pathway was affected in endometrial cancer, and the expression levels of miR-7, miR-17, miR-145, miR-155, miR-206, miR-221, miR-222 were determined. In addition, in silico analyses were examine the molecular interactions between miRNAs and target genes. Identifying dysregulated miRNA expression in endometrial cancer is important for developing miRNA-based therapeutic strategies. In our study, Grade 1 (n = 16), Grade 2 (n = 16), Grade 3 (n = 16), tissues diagnosed with endometrioid adeno carcinoma, control 1 (n = 16) secretory phase and control 2 (n = 16) proliferative phase healthy endometrial tissues without endometrial cancer were included. miRNA expression analysis was performed using the real-time PCR. In our study, the expression of miR-7-5p, miR-145-5p, and miR-206 decreased, whereas the expression of miR-17-5p, miR-221-3p, and miR-222-3p increased in endometrial cancer (p < 0,05). Statistically significant results were not obtained to for the expression levels of miR-21-5p and miR-155-5p. miR-7-5p targets PIK3CD, PIK3R3, PIK3CB and AKT3, miR-17-5p targets PIK3R1 and AKT3, miR-21-5p target PIK3R1, miR-145-5p target AKT3, miR-155-5p targets PIK3CA and PIK3R1, miR-206 target PIK3C2A, miR-221-3p and miR-222-3p target PIK3R1 as identified via in silico analysis. These results can shed light on the development of molecular-targeted therapy strategies. Treatment strategies can be developed by designing ASOs, LNAs, miRNA antagomirs, or miRNA sponges for upregulated miR-17-5p, miR-221-3p, and miR-222-3p, and miRNA mimics for downregulated miR-7-5p, miR-145-5p, and miR-206.
Collapse
Affiliation(s)
- Hasan Öz
- Department of Biochemistry, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Necmiye Canacankatan
- Department of Biochemistry, Faculty of Pharmacy, Mersin University, Mersin, Turkey.
| | - Şerife Efsun Antmen
- Department of Biochemistry, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Hakan Aytan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Ferah Tuncel
- Department of Pathology, Faculty of Medicine, Mersin University, Mersin, Turkey
| |
Collapse
|
265
|
Li J, Wang W, Zhang B, Zhu X, Liu D, Li C, Wang F, Cui S, Ye Z. A clinicoradiological model based on clinical and CT features for preoperative prediction of histological classification in patients with epithelial ovarian cancers: a two-center study. Abdom Radiol (NY) 2025:10.1007/s00261-025-04842-x. [PMID: 39982476 DOI: 10.1007/s00261-025-04842-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/05/2025] [Accepted: 02/09/2025] [Indexed: 02/22/2025]
Abstract
OBJECTIVES To develop and validate a clinicoradiological model integrating clinical and computed tomography (CT) features to preoperative predict histological classification in patients with epithelial ovarian cancers (EOCs). METHODS This retrospective study included 470 patients who were pathologically proven EOCs and performed by contrast enhanced CT before treatment from center I (training cohort, N = 329; internal test cohort, N = 141) and 83 EOC patients who were included as an external test cohort from center II. The univariate analysis and multivariate logistic regression analysis were used to select significant clinical and CT features. The significant clinical model was developed based on clinical characteristics. The significant radiological model was established by CT features. The significant clinical and CT features were used to construct the clinicoradiological model. Model performances were evaluated using the area under the receiver operating characteristic curve (AUC), calibration curve, the Brier score and decision curve analysis (DCA). The AUCs were compared by net reclassification index (NRI) and integrated discrimination improvement (IDI). RESULTS The significant clinical and CT parameters including age, transverse diameter, morphology, margin, ascites and lymphadenopathy were incorporated to build the clinicoradioligical model. The clinicoradiological model showed relatively satisfactory discrimination between type I and type II EOCs with the AUC of 0.841 (95% confidence interval [CI] 0.797-0.886), 0.874 (95% CI 0.811-0.937) and 0.826 (95% CI 0.729-0.923) in the training, internal and external test cohorts, respectively. The NRI and IDI showed the clinicoradiological model significantly performed than those of the clinical model (all P < 0.05). No statistical significance was found between radiological and clinicoradiological model. The clinicoradiological model demonstrated optimal classification accuracy and clinical application value. CONCLUSION The easily accessible nomogram based on the clinicoradiologic model showed favorable performance in distinguishing between type I and type II EOCs and could therefore be used to improve the clinical management of EOC patients.
Collapse
Affiliation(s)
- Jiaojiao Li
- Department of Radiology, First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Wenjiang Wang
- Department of Radiology, First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Bin Zhang
- Department of Radiology, First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Xiaolong Zhu
- Department of Radiology, First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Di Liu
- Department of Radiology, First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Chuangui Li
- Department of Nuclear Medicine, First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Fang Wang
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Shujun Cui
- Department of Radiology, First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| |
Collapse
|
266
|
Mutombo FL, Tsandiraki JK, Mchihiyo T, Wampembe E, Kennedy M, Lodhia J, Chilonga SK. Palliative biliodigestive bypass for unresectable pancreatic malignancy at Kilimanjaro Christian medical centre: a retrospective cross-sectional study. BMC Gastroenterol 2025; 25:103. [PMID: 39984838 PMCID: PMC11846214 DOI: 10.1186/s12876-025-03683-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Pancreatic cancer is a common and deadly cancer, ranking as the 14th most common cancer worldwide and the 7th leading cause of cancer-related deaths. Advanced pancreatic malignancy frequently presents with biliary and gastric outlet obstruction and palliative open interventions are often required, especially in low-income countries where endoscopic surgical bypass methods are often unavailable. This study aimed to describe the demographic and clinical characteristics of patients undergoing biliodigestive bypass for pancreatic malignancy. METHODS This was a hospital-based retrospective observational study at the tertiary hospital in northern Tanzania. We included 53 patients who underwent double or triple bypass surgery for pancreatic malignancy between January 2019 to December 2022 at Kilimanjaro Christian Medical Centre (KCMC), Tanzania. Data was collected from medical records, analyzing demographics, comorbidities, pre-surgery and surgery details, and post-surgery outcomes. Descriptive statistics were used to summarize continuous variables as mean with standard deviation and categorical variables as percentages. RESULTS 53 patients were analyzed, with a mean age of 63.2 years and a male to female ratio of 1.03:1. Jaundice was the most common presentation (77.4%). Of the patients, 74.5% had comorbidities, and the majority (81.1%) were uninsured. 50.9% of patients had a length of hospital stay shorter than 5 days, and 88.3% resumed normal oral intake. Palliation failure was observed in 22.6% of patients. The median survival time for the entire cohort of patients was 65 days. Patients with palliation failure had a significantly shorter mean survival time than those without complications (14.17 vs. 90 days, p = 0.001). CONCLUSION Bypass surgery remains a treatment of choice for palliating symptoms in patients with advanced pancreatic cancer. This study highlights the importance of prompt diagnosis of pancreatic tumors, especially in low-income countries, to achieve better outcomes.
Collapse
Affiliation(s)
- Fabrice Lele Mutombo
- Department of General Surgery, Heal Africa Tertiary Hospital, P.O. Box 319, Goma, Democratic Republic of Congo.
- Department of General Surgery, Kilimanjaro Christian Medical Centre, P.O. Box 3010, Kilimanjaro, Tanzania.
| | - Justin Kambale Tsandiraki
- Department of General Surgery, Heal Africa Tertiary Hospital, P.O. Box 319, Goma, Democratic Republic of Congo
| | - Tumaini Mchihiyo
- Department of General Surgery, Kilimanjaro Christian Medical Centre, P.O. Box 3010, Kilimanjaro, Tanzania
| | - Elizabeth Wampembe
- Department of General Surgery, Kilimanjaro Christian Medical Centre, P.O. Box 3010, Kilimanjaro, Tanzania
| | - Misso Kennedy
- Department of General Surgery, Kilimanjaro Christian Medical Centre, P.O. Box 3010, Kilimanjaro, Tanzania
| | - Jay Lodhia
- Department of General Surgery, Kilimanjaro Christian Medical Centre, P.O. Box 3010, Kilimanjaro, Tanzania
| | - Salum Kondo Chilonga
- Department of General Surgery, Kilimanjaro Christian Medical Centre, P.O. Box 3010, Kilimanjaro, Tanzania
| |
Collapse
|
267
|
Lee SH, Pankaj A, Yilmaz O, Deshpande V, Yilmaz O. Beta-2-microglobulin positive tumor cells and CD8 positive lymphocytes are associated with outcome in post-neoadjuvant colorectal cancer resections. Hum Pathol 2025; 155:105737. [PMID: 39988058 DOI: 10.1016/j.humpath.2025.105737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/13/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Locally advanced colorectal cancers are treated with neoadjuvant therapy (NAT), which has been shown to alter the characteristics of the tumor including size, lymph node yield, and histologic grade. We seek to interrogate the effect of NAT on the immune microenvironment. We compared 190 patients with colorectal adenocarcinoma treated with NAT with those without NAT (n = 926). We evaluated clinicopathologic and molecular factors and performed immunohistochemistry and quantification on tissue microarrays for HLA class I/II proteins, beta-2-microglobulin (B2M), CD8, CD163, LAG3, PD-L1, and FoxP3. Patients in the NAT group were younger (60.9 vs 67.9, p < 0.001) and more often male (59.5 vs. 47.9, p = 0.004) than those in the non-NAT group. Tumors in the NAT group were smaller (3.5 vs 4.7 cm, p < 0.001), less often high grade (6.5% vs. 16.2%, p = 0.001), more frequently in the rectum (68.9% vs. 6.6%, p < 0.001) and associated with lower lymph node yields (p = 0.002); however, the incidence of extramural venous invasion, perineural invasion, and AJCC stage 3-4 disease were not different. Immune cells positive for CD8 (p = 0.011) were significantly lower in the NAT group. A high number of CD8+ cells and higher expression of B2M in tumor cells showed a significant survival benefit in both NAT and non-NAT group. NAT is associated with an immune-low tumor environment. CD8+ cells and tumor B2M expression may help identify a subset of immune high-tumors following NAT. This identification could aid in determining patients who may benefit from conservative management of colorectal carcinomas.
Collapse
Affiliation(s)
- Soo Hyun Lee
- Department of Pathology, Boston Medical Center, Boston, MA, USA
| | - Amaya Pankaj
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Omer Yilmaz
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Vikram Deshpande
- Harvard Medical School, Boston, MA, USA; Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Osman Yilmaz
- Harvard Medical School, Boston, MA, USA; Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
268
|
Lu L, Zhou X, Zheng J, Li D. Coilin Affects the Prognosis of Hepatocellular Carcinoma Through Cell Cycle and Apoptosis. J Hepatocell Carcinoma 2025; 12:367-382. [PMID: 40008396 PMCID: PMC11853879 DOI: 10.2147/jhc.s500119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Purpose Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality with a challenging prognosis. HCC lacks effective prognostic biomarkers. We investigated the diagnostic and prognostic value of COIL expression in HCC. Patients and Methods This study evaluated the expression and prognostic significance of COIL using data from the TCGA and local hospital samples, with 374 and 118 liver cancer patients in the TCGA database and local hospital, respectively. The techniques include bioinformatics analysis, qRT-PCR, immunohistochemistry (IHC), and in vitro cell experiments, which encompass CCK-8 assays, wound healing assays, and Transwell invasion assays. The relationship between COIL expression and clinical outcomes was assessed, and COIL's biological function in HCC was investigated through cellular assays. Results Analysis of cell lines and HCC tissue samples revealed that COIL mRNA or protein expression levels were significantly higher in HCC cell lines/tissues compared to normal liver cells/tissues. Univariate and multivariate analyses indicated that COIL is an independent prognostic factor for overall survival (OS) in HCC. Additionally, 14% of HCC patients had alterations in the COIL gene, and patients with COIL gene alterations had significantly lower OS (p<0.001) and disease-free survival (DFS) (p<0.001) compared to those without gene alterations. Knockdown of COIL expression inhibited the proliferation, migration, and invasion of Hep3B, HepG2, and Huh7. Compared to the control group, COIL knockdown cells showed a marked reduction in CDC25C and CCNB1 protein levels, suggesting that COIL knockdown leads to G2/M phase cell cycle arrest. After COIL knockdown, caspase-3 and BCL-2 protein levels were downregulated, while cleaved caspase and BAX protein levels were upregulated, indicating that COIL knockdown promotes apoptosis in liver cancer cells. Conclusion COIL is an independent predictor of prognosis. COIL's association with poor OS and its role in enhancing cancer cell proliferation and invasion highlight its potential as a therapeutic target.
Collapse
Affiliation(s)
- Lingling Lu
- Department of Infection Disease, Fujian Medical University Union Hospital, Fuzhou, People’s Republic of China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, People’s Republic of China
| | - Xiaoling Zhou
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Hepatobiliary Disease, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, People’s Republic of China
| | - Jiaolong Zheng
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Hepatobiliary Disease, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, People’s Republic of China
| | - Dongliang Li
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Hepatobiliary Disease, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, People’s Republic of China
| |
Collapse
|
269
|
Marchese U, Pauly V, Pellat A, Richa Y, Fond G, Tzedakis S, Gaillard M, Fuchs B, Orleans V, Fuks D, El Amrani M, Boyer L. End-of-life care for patients with pancreatic cancer in France: a nationwide population-based cohort study. Ther Adv Med Oncol 2025; 17:17588359251320731. [PMID: 39990013 PMCID: PMC11843702 DOI: 10.1177/17588359251320731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
Background Pancreatic cancer, a frequently fatal disease with severe symptoms, can require high-intensity end-of-life (HI-EOL) care, posing challenges to patients' well-being. The examination of HI-EOL care to develop tailored interventions in the management of pancreatic cancer is a critical, yet underexplored area. Objectives The objective of this study was to assess the factors that influence the intensity of end-of-life (EOL) care in France. Design A retrospective study of patients registered in the French Nationwide database who were hospitalized in France for pancreatic adenocarcinoma from January 1, 2014 to December 31, 2019, and subsequently died during the follow-up period. Methods Data on patient demographics, clinical characteristics, hospitalization details, and palliative care were collected. The primary outcome measure was the evaluation of HI-EOL care, defined by indicators such as death in an intensive care unit (ICU), multiple hospitalizations, and chemotherapy administration within the last 30 days of life. Secondary outcomes included indicators of most-intensive EOL (MI-EOL) care and invasive procedures (IP). Univariate and multivariate logistic regression analyses were conducted to identify factors associated with each outcome measure. Results A total of 42,696 patients who died from pancreatic adenocarcinoma were included. Among them, 41.1% experienced HI-EOL, with the most common indicators being multiple hospitalizations and death in an ICU, emergency room, or acute care unit. A smaller proportion (2.8%) received MI-EOL care, while 28.1% underwent IPs in the last 30 days of life. The multivariate analysis revealed that male gender and follow-up in non-cancer specialized care facilities were associated with a higher risk of HI-EOL. Conversely, palliative care involvement and older age at death were identified as protective factors. Male gender, older age at death, and palliative care involvement were associated with lower rates of MI-EOL care and IPs. Conclusion These results underscore the importance of palliative care integration and individualized approaches in improving the EOL quality of care and patient outcomes for individuals with advanced pancreatic cancer.
Collapse
Affiliation(s)
- Ugo Marchese
- Department of Digestive, HPB and Endocrine Surgery, Hôpital Cochin, AP-HP Centre, 27 rue du Faubourg St Jacques, Paris 75014, France
- Université de Paris Cité, 15 rue de l’école de médecine 75006 Paris, France
| | - Vanessa Pauly
- Département d’Information Médicale, Hôpital de la Timone, 264 Rue Saint-Pierre, 13005 Marseille, France
- Université d’Aix-Marseille, Jardin du Pharo, 58 Boulevard Charles Livon, 13007 Marseille, France
| | - Anna Pellat
- Department of Gastroenterology and Digestive Oncology, Hôpital Cochin, AP-HP Centre, 27 rue du Faubourg St Jacques, 75014 Paris, France
- Université de Paris Cité, 15 rue de l’école de médecine 75006 Paris, France
| | - Yasmina Richa
- School of Medicine, University College Cork, Cork, Ireland
| | - Guillaume Fond
- Department of University Psychiatry, Sainte Marguerite University Hospital, Assistance Publique des Hôpitaux de Marseille, Marseille, France
- Assistance Publique des Hôpitaux de Marseille, Aix-Marseille University, UR3279: Health Service Research and Quality of Life Center - CEReSS, Marseille, France
| | - Stylianos Tzedakis
- Department of Digestive, HPB and Endocrine Surgery, Hôpital Cochin, AP-HP Centre, 27 rue du Faubourg St Jacques, 75014 Paris, France
- Université de Paris Cité, 15 rue de l’école de médecine 75006 Paris, France
| | - Martin Gaillard
- Department of Digestive, HPB and Endocrine Surgery, Hôpital Cochin, AP-HP Centre, 27 rue du Faubourg St Jacques, 75014 Paris, France
- Université de Paris Cité, 15 rue de l’école de médecine 75006 Paris, France
| | - Basile Fuchs
- Département d’Information Médicale, Hôpital de Brest, 2 avenue Foch, 29200 Brest, France
| | - Veronica Orleans
- Département d’Information Médicale, Hôpital de la Timone, 264 Rue Saint-Pierre, 13005 Marseille, France
- Université d’Aix-Marseille, Jardin du Pharo, 58 Boulevard Charles Livon, 13007 Marseille, France
| | - David Fuks
- Department of Digestive Surgery, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland
| | - Mehdi El Amrani
- Department of Digestive Surgery and Transplantation, CHRU de Lille, 2 Av. Oscar Lambret, 59000 Lille, France
- Université de Lille, 42 Rue Paul Duez, 59000 Lille, France
| | - Laurent Boyer
- Département d’Information Médicale, Hôpital de la Timone, 264 Rue Saint-Pierre, 13005 Marseille, France
- Université d’Aix-Marseille, Jardin du Pharo, 58 Boulevard Charles Livon, 13007 Marseille, France
| |
Collapse
|
270
|
Li J, Liu J, Wu Y, Sun Y, Huang G, Jin M. α-Hederin inhibited pancreatic cancer cell malignant progression by inhibiting LDHA-mediated glycolysis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03621-7. [PMID: 39969605 DOI: 10.1007/s00210-024-03621-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/08/2024] [Indexed: 02/20/2025]
Abstract
α-Hederin is a pentacyclic triterpenoid saponin extracted from Pulsatilla chinensis, which is known to suppress cancer cell proliferation. However, the role of this compound in pancreatic cancer cells remains unclear. The aim of this study was to reveal the docking molecular and the regulatory mechanism of α-hederin in pancreatic cancer. Here, we cultured Capan-1 and BxPC-3 cells and treated with different doses of α-hederin. Cell proliferation, migration, and apoptosis were detected using CCK8, EdU, Transwell, wound healing assay, and flow cytometer apoptosis assay. The in vivo experiment using subcutaneous tumor and caudal vein metastasis model to evaluate the inhibit effect of α-hederin Capan-1 cell tumor growth and metastasis. Proteomics were used to reveal the regulatory mechanism. The result shows that α-hederin treatment inhibits cell proliferation and invasion in concentration dependence way in both vivo and in vitro. The result shows that the IC50 for both Capan-1 and BxPC-3 were 32.5 Mµ and 15 Mµ, respectively. Flow cytometer apoptosis assay shows that α-hederin treatment promotion cell apoptosis in both Capan-1 and BxPC-3 cells. Proteomics and immunofluorescence detection confirmed that α-hederin treatment downregulated lactate dehydrogenase A (LDHA) expression and inhibited glycolysis. Molecular docking of α-hederin and LDHA proteins further confirmed that LDHA is a target of α-hederin. Taken together, this study confirms that α-hederin inhibits pancreatic cancer cell proliferation and invasion by inhibiting LDHA-mediated glycolysis. LDHA may be a direct target of α-hederin in pancreatic cancer.
Collapse
Affiliation(s)
- Jingjing Li
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Jiao Liu
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Yue Wu
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Yi Sun
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, No.1111, XianXia Road, Shanghai, 200336, China.
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| | - Mingming Jin
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| |
Collapse
|
271
|
Zhou X, Ba Y, Xu N, Xu H, Zhang Y, Liu L, Weng S, Liu S, Xing Z, Chen S, Luo P, Wang L, Han X. Pharmacogenomics-based subtype decoded implications for risk stratification and immunotherapy in pancreatic adenocarcinoma. Mol Med 2025; 31:62. [PMID: 39972282 PMCID: PMC11837470 DOI: 10.1186/s10020-024-01049-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 12/16/2024] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND With fatal malignant peculiarities and poor survival rate, outcomes of pancreatic adenocarcinoma (PAAD) were frustrated by non-response and even resistance to therapy due to heterogeneity across clinical patients. Nevertheless, pharmacogenomics has been developed for individualized-treatment and still maintains obscure in PAAD. METHODS A total of 964 samples from 10 independent multi-center cohorts were enrolled in our study. With drug response data from the profiling of relative inhibition simultaneously in mixtures (PRISM) and genomics of drug sensitivity in cancer (GDSC) databases, we established and validated multidimensionally three pharmacogenomics-classified subtypes using non-negative matrix factorization (NMF) and nearest template prediction (NTP) algorithms, separately. The heterogenous biological characteristics and precision medicine strategies among subtypes were further investigated. RESULTS Three pharmacogenomics-classified subtypes after stable and reproducible validation, distinguished in six aspects of prognosis, biological peculiarities, immune landscapes, genomic variations, immunotherapy and individualized management strategies. Subtype 2 was close to immunocompetent phenotype and projected to immunotherapy; Subtype 3 held most favorable outcomes and metabolic pathways distinctively, promising to be treated with first-line agents. Subtype 1 with worst prognosis, was anticipated to chromosome instability (CIN) phenotype and resistant to chemotherapeutic agents. In addition, ITGB6 contributed to subtype 1 resistance to 5-fluorouracil, and knockdown of ITGB6 enhanced sensitivity to 5-fluorouracil in in vitro experiments. Ultimately, appropriate clinical stratified treatments were assigned to corresponding subtypes according to pharmacogenomic transcripts. Some limitations were not taken into account, thus needs to be supported by more research. CONCLUSION A span-new molecular subtype exploited for PAAD uncovered an insight into precise medication on ground of pharmacogenomics, and highly refined multiple clinical management strategies for specific patients.
Collapse
Affiliation(s)
- Xing Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Nuo Xu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Long Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shutong Liu
- School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, China
| | - Zhe Xing
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuang Chen
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Libo Wang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China.
| |
Collapse
|
272
|
Dong S, Li X, Chen Z, Shi H, Wang Z, Zhou W. MMP28 recruits M2-type tumor-associated macrophages through MAPK/JNK signaling pathway-dependent cytokine secretion to promote the malignant progression of pancreatic cancer. J Exp Clin Cancer Res 2025; 44:60. [PMID: 39972459 PMCID: PMC11837641 DOI: 10.1186/s13046-025-03321-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/06/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Crosstalk between pancreatic cancer cells and tumor-associated macrophages (TAMs) is a critical driver of malignant progression, and plays an important role in the low response rate to immunotherapy in patients with for pancreatic cancer. Although it is known that cancer cells induce TAM infiltration and M2 polarization, the underlying mechanisms remain elusive. Herein, we identified matrix metalloproteinase 28 (MMP28), a highly expressed protein, as a key regulator of this process. METHODS Immunohistochemical staining and qRT-PCR were used to validate MMP28 as a potential marker for the prognosis of patients with pancreatic cancer. We evaluated the tumor-promoting effect of MMP28 in vitro with CCK-8, Transwell, and EdU assay and Western blotting and explored the potential mechanism of MMP28-induced M2 polarization of TAMs with a coculture system, immunofluorescence staining and flow cytometry. A subcutaneous graft tumor model was constructed to assess the tumor-promoting effect of MMP28 and its ability to induce M2 TAM infiltration. RESULTS The relevant results of this study revealed a strong correlation between MMP28 expression and TAM infiltration, with a predominance of M2-polarized TAMs in pancreatic cancer tissues. Mechanistic investigations demonstrated that MMP28 promotes the secretion of multiple cytokines, including IL-8 and VEGFA through the activation of the MAPK/JNK signaling pathway. These cytokines act as potent chemoattractants and polarizing factors for TAMs. Additionally, we discovered an interaction between MMP28 and ANXA2, which contributes to the regulation of TAM recruitment and polarization. In vivo studies confirmed the critical role of MMP28 in tumor growth and TAM infiltration. Depletion of macrophages, inhibition of JNK, or neutralization of IL-8 and VEGFA significantly suppressed tumor progression. Transcriptomic analysis suggested that IL-8 and VEGFA induce M2 TAM polarization by modulating TAM amino acid metabolism. CONCLUSIONS Collectively, our findings elucidate a novel mechanism by which pancreatic cancer cells manipulate the tumor microenvironment through MMP28-dependent cytokine secretion, promoting TAM infiltration and M2 polarization. These results highlight MMP28 as a promising therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Shi Dong
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| | - Xin Li
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| | - Zhou Chen
- Department of Thoracic Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Huaqing Shi
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| | - Zhengfeng Wang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| | - Wence Zhou
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China.
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
273
|
Pasaol JC, Śmieszek A, Pawlak A. Exploring the Therapeutic Potential of BRCA1 and BRCA2 as Targets in Canine Oncology: A Comprehensive Review of Their Role in Cancer Development and Treatment. Int J Mol Sci 2025; 26:1768. [PMID: 40004231 PMCID: PMC11855874 DOI: 10.3390/ijms26041768] [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: 01/15/2025] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor diseases represent a significant global health challenge, impacting both humans and companion animals, notably dogs. The parallels observed in the pathophysiology of cancer between humans and dogs underscore the importance of advancing comparative oncology and translational research methodologies. Furthermore, dogs serve as valuable models for human cancer research due to shared environments, genetics, and treatment responses. In particular, breast cancer gene 1 (BRCA1) and breast cancer gene 2 (BRCA2), which are critical in human cancer, also influence the development and progression of canine tumors. The role of BRCA1 and BRCA2 in canine cancers remains underexplored, but its potential significance as therapeutic targets is strongly considered. This systematic review aims to broaden the discussion of BRCA1 and BRCA2 beyond mammary tumors, exploring their implications in various canine cancers. By emphasizing the shared genetic underpinnings between species and advocating for a comparative approach, the review indicates the potential of BRCA genes as targets for innovative cancer therapies in dogs, contributing to advances in human and veterinary oncology.
Collapse
Affiliation(s)
| | | | - Aleksandra Pawlak
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Norwida 31, 50-375 Wrocław, Poland; (J.C.P.); (A.Ś.)
| |
Collapse
|
274
|
Reymova F, Sever B, Topalan E, Sevimli-Gur C, Can M, Tuyun AF, Başoğlu F, Ece A, Otsuka M, Fujita M, Demirci H, Ciftci H. Design, Synthesis, and Mechanistic Anticancer Evaluation of New Pyrimidine-Tethered Compounds. Pharmaceuticals (Basel) 2025; 18:270. [PMID: 40006082 PMCID: PMC11859636 DOI: 10.3390/ph18020270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/12/2025] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Despite recent breakthroughs in cancer treatment, non-small cell lung cancer (NSCLC) and breast cancer remain major causes of death from all malignancies. The epidermal growth factor receptor (EGFR) is an important mediator of the pathways involved in cell proliferation, apoptosis, and angiogenesis. Thus, its overexpression triggers several types of cancer, including NSCLC and breast cancer. Methods: In the current study, we synthesized new pyrimidine-tethered compounds (chalcone derivative (B-4), pyrazoline-carbothioamide (B-9), and pyrazoline-thiazole hybrids (BH1-7)). These compounds were then tested for cytotoxicity against A549 NSCLC and MCF-7 breast cancer cells. Results: Of these, B-4 displayed significant cytotoxicity against both cells (IC50 = 6.70 ± 1.02 µM for MCF-7; IC50 = 20.49 ± 2.7 µM for A549) compared to the standard agent lapatinib (IC50 = 9.71 ± 1.12 µM for MCF-7; IC50 = 18.21 ± 3.25 µM for A549). The anticancer potential of B-4 between Jurkat leukemic T cells and peripheral blood mononuclear cells (PBMCs) (healthy) was found to be selective. Mechanistically, 11.9% and 10.2% of A549 and MCF-7 cells treated with B-4, respectively, underwent apoptosis and B-4 produced 46% EGFR inhibition at a concentration of 10 μM. The B-4/EGFR complex obtained after induced fit docking was subjected to 300 ns of molecular dynamics simulation, which confirmed the stability of the complex in a mimicked biological environment. On the other hand, B-4 was shown to have drug-like properties by in silico pharmacokinetic estimation. Conclusions: B-4 is an EGFR inhibitor and apoptosis inducer for future NSCLC and breast cancer studies.
Collapse
Affiliation(s)
- Farida Reymova
- Department of Bioengineering Sciences, Izmir Katip Celebi University, Izmir 35620, Türkiye; (F.R.); (M.C.)
| | - Belgin Sever
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Türkiye;
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan; (M.O.); (M.F.)
| | - Edanur Topalan
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Türkiye; (E.T.); (H.D.)
| | - Canan Sevimli-Gur
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir 35620, Türkiye;
| | - Mustafa Can
- Department of Bioengineering Sciences, Izmir Katip Celebi University, Izmir 35620, Türkiye; (F.R.); (M.C.)
| | - Amaç Fatih Tuyun
- Department of Chemistry, Faculty of Science, Istanbul University, Fatih, Istanbul 34126, Türkiye;
| | - Faika Başoğlu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, European University of Lefke, Northern Cyprus, TR-10, Mersin 99800, Türkiye;
| | - Abdulilah Ece
- Department of Medical Biochemistry, Faculty of Medicine, Biruni University, İstanbul 34015, Türkiye;
| | - Masami Otsuka
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan; (M.O.); (M.F.)
- Department of Drug Discovery, Science Farm Ltd., Kumamoto 862-0976, Japan
| | - Mikako Fujita
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan; (M.O.); (M.F.)
| | - Hasan Demirci
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Türkiye; (E.T.); (H.D.)
| | - Halilibrahim Ciftci
- Department of Bioengineering Sciences, Izmir Katip Celebi University, Izmir 35620, Türkiye; (F.R.); (M.C.)
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan; (M.O.); (M.F.)
- Department of Drug Discovery, Science Farm Ltd., Kumamoto 862-0976, Japan
- Department of Molecular Biology and Genetics, Burdur Mehmet Akif Ersoy University, Istiklal Campus, Burdur 15030, Türkiye
| |
Collapse
|
275
|
Alqudah MAY, Yaseen MM, Alzoubi KH, Al-Husein BA, Bardaweel SK, Abuhelwa AY, Semreen AM, Zenati RA, El-Awady R, Shara M, Bustanji Y, Soares NC, Abu-Gharbieh E, Ramadan WS, Semreen MH. Metabolomic Analysis, Antiproliferative, Anti-Migratory, and Anti-Invasive Potential of Amlodipine in Lung Cancer Cells. Drug Des Devel Ther 2025; 19:1215-1229. [PMID: 39991087 PMCID: PMC11847429 DOI: 10.2147/dddt.s484561] [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: 06/27/2024] [Accepted: 02/06/2025] [Indexed: 02/25/2025] Open
Abstract
Background and Objective Lung cancer stands as the leading cause of cancer-related fatalities worldwide. While chemotherapy remains a crucial treatment option for managing lung cancer in both early-stage and advanced cases, it is accompanied by significant drawbacks, including severe side effects and the development of chemoresistance. Overcoming chemoresistance represents a considerable challenge in lung cancer treatment. Amlodipine cytotoxicity was previously demonstrated and could make lung cancer cells more susceptible to chemotherapies. This research aims to examine the metabolomics changes that may occur due to amlodipine's anticancer effects on non-small cell lung cancer (NSCLC) cells. Methods Amlodipine's effects on A549 and H1299 NSCLC were evaluated using a colorimetric MTT assay, a scratch wound-healing assay and Matrigel invasion chambers to measure cell viability, cell migration and cell invasion. Ultra-high-performance liquid chromatography-electrospray ionization quadrupole time-of-flight mass spectrometry (UHPLC-ESI-QTOF-MS) was used for the untargeted metabolomics investigation. Results Our study revealed that amlodipine significantly reduced proliferation of cancer cells in a dose-dependent fashion with IC50 values of 23 and 25.66 µM in A549 and H1299 cells, respectively. Furthermore, amlodipine reduced the invasiveness and migration of cancer cells. Metabolomics analysis revealed distinct metabolites to be significantly dysregulated (Citramalic acid, L-Proline, dGMP, L-Glutamic acid, Niacinamide, and L-Acetylcarnitine) in amlodipine-treated cells. Conclusion The present study illustrates the anticancer effects of amlodipine on lung cancer proliferation, migration, and invasion in vitro and enhance our understanding of how amlodipine exerts its anticancer potential by casting light on these mechanisms.
Collapse
Affiliation(s)
- Mohammad A Y Alqudah
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Mahmoud M Yaseen
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Belal A Al-Husein
- Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Sanaa K Bardaweel
- Department of Pharmaceutical Sciences, School of Pharmacy, the University of Jordan, Amman, Jordan
| | - Ahmad Y Abuhelwa
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, United Arab Emirates
| | - Ahlam M Semreen
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ruba A Zenati
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Raafat El-Awady
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Mohd Shara
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, United Arab Emirates
| | - Yasser Bustanji
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, the University of Jordan, Amman, Jordan
| | - Nelson C Soares
- Department of Medicinal Chemistry, University of Sharjah, Sharjah, United Arab Emirates
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University Medicine and Health Sciences (MBRU), Dubai Health, Dubai, United Arab Emirates
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai Health, Dubai, United Arab Emirates
| | - Eman Abu-Gharbieh
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, the University of Jordan, Amman, Jordan
| | - Wafaa S Ramadan
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Mohammad H Semreen
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
276
|
Pflug C, Müller F, Koseki JC, Petersen C, Nienstedt JC, Tribius S. Objective dysphagia is very common after radiotherapy in oropharyngeal cancer patients. Oral Surg Oral Med Oral Pathol Oral Radiol 2025:S2212-4403(25)00779-5. [PMID: 40169338 DOI: 10.1016/j.oooo.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/30/2024] [Accepted: 02/08/2025] [Indexed: 04/03/2025]
Abstract
OBJECTIVE Dysphagia is one of the most serious adverse events in the treatment of head and neck cancer. This cross-sectional study aimed to assess pharyngeal residue, and penetration/aspiration in oropharyngeal cancer patients (OPC) after radiotherapy using flexible endoscopic evaluation of swallowing (FEES). METHODS A total of 35 OPC patients who had received radio-(chemo) therapy (R(C)T), including 8 patients with primary R(C)T), were included and examined by FEES to determine the swallowing status and were asked to indicate their swallowing ability on a visual scale to reflect the problem perceived by the patient. During FEES the patients were given three standardized bolus consistencies and four test pills. Penetration, aspiration, and residue were evaluated and classified. RESULT Relevant dysphagia was present in 23/35 (66%) patients. Almost half of all patients (15/35) showed aspiration (53% (8/15) silent). Residue occurred in 91% but without correlation to aspiration. A significant association between dysphagia and impaired pill swallowing was found (P = .003) occurring in 20 of 35 patients. Even in patients with small tumors and without prior surgery severe dysphagia was found. CONCLUSIONS Severe dysphagia is frequent after R(C)T affecting more than half of the patients with OPC. The frequent impaired pill swallowing ability should be considered Therefore, regular dysphagia diagnostics in the follow-up setting are advisable to initiate appropriate treatment and raise patients' quality of life, prevent aspiration pneumonia, and improve overall outcomes after tumor therapy.
Collapse
Affiliation(s)
- Christina Pflug
- Department of Voice, Speech and Hearing Disorders, Center for Clinical Neurosciences, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Frank Müller
- Department of Voice, Speech and Hearing Disorders, Center for Clinical Neurosciences, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana-Christiane Koseki
- Department of Voice, Speech and Hearing Disorders, Center for Clinical Neurosciences, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cordula Petersen
- Department of Radiotherapy and Radiation Oncology, Center for Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julie Cläre Nienstedt
- Department of Voice, Speech and Hearing Disorders, Center for Clinical Neurosciences, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Silke Tribius
- Department of Radiation Oncology, Asklepios Hospital St. Georg, Hamburg, Germany; Hermann-Holthusen Institute for Radiation Oncology, Asklepios Hospital St. Georg, Hamburg, Germany
| |
Collapse
|
277
|
Liu X, Liu J, Zeng Y, Qiao D, Wang Q. AL365181.3 as a novel prognostic biomarker for lung adenocarcinoma. Sci Rep 2025; 15:5853. [PMID: 39966648 PMCID: PMC11836369 DOI: 10.1038/s41598-025-90008-0] [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: 08/04/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
As a lncRNA, AL365181.3 is aberrantly expressed in multiple cancer types, including lung adenocarcinoma (LUAD). However, the biological process underlying the ability of AL365181.3 to promote the progression of LUAD is unclear. Here, the pancancer expression level of AL365181.3 was analyzed via the TCGA and GTEx databases, as well as its clinical characteristics and prognostic value. We investigated the biological functions of AL365181.3 in LUAD using various in vitro and in vivo assays. We found that AL365181.3 was significantly more highly expressed in many types of cancer tissues, including LUAD tissues, than in adjacent normal tissues. LUAD patients with high AL365181.3 expression had poor prognoses. Functional enrichment analyses indicated that AL365181.3 is involved in the regulation of metabolism, MAPK signaling and other tumor regulatory signaling pathways. Finally, we found that AL365181.3 knockdown reduced the proliferation, migration and invasion capacity of LUAD cells, and AL365181.3 knockdown resulted in a reduced in vivo tumorigenic capacity of LUAD cells. These findings provide a comprehensive understanding of the role of AL365181.3 in LUAD.
Collapse
Affiliation(s)
- Xiaoying Liu
- Xinxiang Medical University, Xinxiang, 453003, Henan, China
- Department of Thoracic Surgery, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong New Area, Shanghai, 201318, China
| | - Jinlong Liu
- Xinxiang Medical University, Xinxiang, 453003, Henan, China
- Department of Thoracic Surgery, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong New Area, Shanghai, 201318, China
| | - Yingou Zeng
- Department of Thoracic Surgery, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong New Area, Shanghai, 201318, China
| | - Di Qiao
- Department of Thoracic Surgery, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong New Area, Shanghai, 201318, China
| | - Qiang Wang
- Department of Thoracic Surgery, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong New Area, Shanghai, 201318, China.
| |
Collapse
|
278
|
Boyland R, Amin S, Shostrom V, Zheng C, Allison J, Lin C. Comparison of overall survival of adult and pediatric osteosarcoma patients using the national cancer database. BMC Cancer 2025; 25:290. [PMID: 39966776 PMCID: PMC11837598 DOI: 10.1186/s12885-025-13496-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/12/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Differences in overall survival (OS) between pediatric and adult patients diagnosed with osteosarcoma are poorly understood. The objective of this study is to compare the OS of pediatric and adult patients with osteosarcoma, identify prognostic factors associated with OS, and explore factors specifically associated with pediatric osteosarcoma using data gathered from the National Cancer Database (NCDB). METHODS Patients > = 1 years old and diagnosed with osteosarcoma between 2004 and 2017 were included in the study. Multivariable Cox regression analysis adjusted for gender, race, income, education, place of living, health insurance status, year of diagnosis, stage of cancer, surgery, chemotherapy, and radiation therapy (RT) was used to assess the association of age with the OS of the patients. RESULTS The analysis included 8,458 patients among whom 3,027 (35.8%) were between 1 and 17 years old. In the multivariable Cox regression analysis, adult patients had worse OS compared with pediatric patients (HR: 1.84; p < .01). When stratified by treatment type, pediatric patients had better OS in several groups. This includes those who received chemotherapy alone (HR: 0.58, p < .01), surgery alone (HR: 0.48, p < .01), surgery plus chemotherapy (HR: 0.56, p < .01), and those who received no treatment (HR: 0.31, p < .01). There was no significant difference in OS between pediatric and adult patients receiving a combination of chemotherapy, surgery, and RT (HR: 0.81, p = .42). In analysis stratified by cancer stage, pediatric patients had better OS compared to adult patients at each stage. Multivariable logistic regression analysis revealed that pediatric patients are more likely to be non-white, have insurance, present with unknown/occult stage disease, have poorly differentiated tumors, and receive chemotherapy, or surgery. Additionally, multivariable Cox regression analysis identified factors associated with improved OS: age, diagnosis between 2011 and 2015, private insurance, non-metastatic disease, well-differentiated tumors, and receiving chemotherapy or surgery, but not RT. CONCLUSION Pediatric patients diagnosed with osteosarcoma had better OS compared to their adult counterparts. Pediatric patients had better OS compared to adults when the analysis was stratified by treatment modality and stage of cancer. Further research is necessary to elucidate the underlying reason for this difference.
Collapse
Affiliation(s)
- Ryan Boyland
- Department of Emergency Medicine, Denver Health Medical Center, Denver, CO, USA
| | - Saber Amin
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Valerie Shostrom
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Cheng Zheng
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jenna Allison
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Chi Lin
- Department of Emergency Medicine, Denver Health Medical Center, Denver, CO, USA.
- Department of Radiation Oncology, University of Nebraska Medical Center, 986861 Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
279
|
Shen Y, Dong X, Li X, Shi Z, Shao T, Jiang J, Song J. WNT inhibitor SP5-mediated SERPING1 suppresses lung adenocarcinoma progression via TSC2/mTOR pathway. Cell Death Dis 2025; 16:103. [PMID: 39962118 PMCID: PMC11832940 DOI: 10.1038/s41419-025-07440-3] [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: 10/03/2024] [Revised: 01/25/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
The long-term outlook for patients grappling with lung cancer (LC) remains bleak, with lung adenocarcinoma (LUAD) emerging as the most predominant histological subtype. Our Mendelian randomization (MR) investigation spotlighted that heightened levels of the circulating protein serpin peptidase inhibitor family G1 (SERPING1) substantially mitigated LC risk. The fusion of multi-omics strategies unveiled that SERPING1 exhibited diminished expression in LUAD patients compared to healthy individuals both in tissues and serum, with LUAD individuals showcasing elevated SERPING1 expression demonstrating improved prognoses. Furthermore, SERPING1 expression exhibited a robust correlation with the efficacy of immunotherapy. Through meticulous in vivo and in vitro analyses, we unraveled that SERPING1 impeded the proliferation, migration, invasion and wound healing of LUAD cells via the tuberous sclerosis 2 (TSC2)/mammalian target of rapamycin (mTOR) pathway. Mechanistically, WNT inhibitor- Specificity Protein (SP5) was delineated as facilitator of SERPING1 transcription by binding to the SERPING1 gene promoter. Intriguingly, aside from the association between SERPING1 and systolic blood pressure, glycosylated hemoglobin (HbA1c), type I diabetes, no discernible link between SERPING1 overexpression and heightened risks of other cardiometabolic conditions and diseases was evident. In summary, SERPING1 emerges as a novel tumor suppressor gene and SP5/SERPING1/TSC2 is a promising therapeutic target in the context of LUAD.
Collapse
Affiliation(s)
- Yefeng Shen
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences, Nanning, China
- Department of Thoracic Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaofeng Dong
- Department of Hepatobiliary, Pancreas and Spleen Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region (Guangxi Academy of Medical Sciences), Nanning, China
| | - Xujia Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhiyuan Shi
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Tingting Shao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Junlan Jiang
- Department of Pathology, the First Affiliated Hospital, Anhui Medical University, Hefei, China
- Pathology Center, Anhui Medical University, Hefei, China
| | - Jian Song
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences, Nanning, China.
- Department of Radiation Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
280
|
Bose S, Sharma S, Kumar A, Mishra Y, Mishra V. Ovarian cancer and its management through advanced drug delivery system. Med Oncol 2025; 42:76. [PMID: 39960609 DOI: 10.1007/s12032-025-02621-8] [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: 11/09/2024] [Accepted: 01/29/2025] [Indexed: 02/28/2025]
Abstract
Ovarian cancer is the deadliest gynecological cancer, often being diagnosed at a late-stage when the disease is already advanced. This makes it known as the ''silent killer'' due to its high mortality rate among women. The ovary itself is composed of three main types of cells epithelial cells, germ cells, and stromal cells. Over 90% of all occurrences of ovarian cancer are epithelial ovarian carcinoma. Ovarian cancer is responsible for 2.5% of cancer in women. The primary signs include stomach bloating, trouble eating or feeling full rapidly, fatigue, and discomfort during intercourse. The management of ovarian cancer is improved by advanced drug delivery systems because they increase therapeutic targeting, reduce side effects, and overcome drug resistance. Accurate distribution to cancer cells is made possible by methods such as ligand-functionalized nanomedicines, dual-targeted nano-drugs, drug conjugates, antibody-drug conjugates and peptide/folate drug conjugates. These technologies enhance individualized and successful ovarian cancer treatment outcomes by maximizing chemotherapy efficacy, extending drug release, and lowering toxicity.
Collapse
Affiliation(s)
- Sujit Bose
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| | - Shubham Sharma
- School of Pharmaceutical Sciences, CT University, Ludhiana, Punjab, 142024, India
| | - Atul Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Yachana Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| |
Collapse
|
281
|
Jamil A, Qureshi Z, Siddique R, Altaf F, Jamil R, Wali N. A Meta-analysis on Effects of Chimeric Antigen Receptor T-cell Therapy in Relapsed or Refractory B-cell Acute Lymphoblastic Leukemia. Am J Clin Oncol 2025:00000421-990000000-00258. [PMID: 39956997 DOI: 10.1097/coc.0000000000001176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
OBJECTIVES This review evaluates the long-term outcomes and adverse events associated with chimeric antigen receptor (CAR) T-cell therapy in patients with relapsed/refractory B-cell acute lymphoblastic leukemia (r/r B-ALL). METHODS We conducted the search in relevant databases up to June 2024. We included clinical trials on CAR T-cell therapy for patients with r/r B-ALL. Meta-analyses were conducted using Comprehensive Meta-Analysis V3 and Review Manager 5.4. RESULTS Out of 2659 identified studies, 10 were included in this review. The pooled analysis demonstrated a high minimal residual disease-negative complete remission, with an overall event rate (ER) of 70% (95% CI: 61%-78%, I2 =8 8.35%). Anti-CD19 CAR T-cell therapy showed the highest efficacy with an ER of 74.75% (95% CI: 61%-80%, I2 = 89.84%). Combination therapies targeting CD19 and CD22 had an ER of 69% (95% CI: 53%-83%, I2 = 82.56%). Significant adverse effects included cytokine release syndrome with a mean incidence of 81.8% (95% CI: 76.7%-86.9%), neurotoxicity at 33.2% (95% CI: 28.1%-38.3%), and hematologic toxicities at 71.9% (95% CI: 66.4%-77.4%). CONCLUSIONS CAR T-cell therapy is a groundbreaking advancement in treating r/r B-ALL, offering high rates of durable remissions.
Collapse
Affiliation(s)
- Abdur Jamil
- Department of Medicine, Samaritan Medical Centre
| | - Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, CT
| | | | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY
| | | | - Neehal Wali
- Vituity Hospitalist Group, HSHS St. John's Hospital Springfield, IL
| |
Collapse
|
282
|
Moeng S, Chamorro-Parejo AD, Jeon MS, Cai JJ, Ramos KS. Single-Cell RNA Sequencing Reveals Extensive Heterogeneity and Unique Gene Trajectories in Non-Transformed and Transformed Human Lung Epithelial Cells: Insights into the Role of LncRNAs in Tumor Heterogeneity. Int J Mol Sci 2025; 26:1690. [PMID: 40004153 PMCID: PMC11855061 DOI: 10.3390/ijms26041690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Lung cancer exhibits substantial inter- and intra-tumor heterogeneity, with features that present significant challenges in advancing biomarker discovery and the development of targeted therapeutics. To fill this gap, we employed single-cell RNA sequencing (scRNA-seq) and advanced bioinformatics tools to evaluate the transcriptomic heterogeneity of immortalized, non-transformed (BEAS2B) and transformed (H460) lung epithelial cell lines and their responses to carcinogen challenge. Gene expression profiles resolved four primary clusters further discretized into unique subclusters based on genetic signatures and phenotypic profiles. Profiles of long non-coding RNAs (lncRNAs) identified microRNA host genes, antisense RNA genes, divergent transcript, and long intergenic non-coding RNAs as contributors to cellular heterogeneity. These findings indicate that distinct patterns of gene expression, remarkably in lncRNAs, define cellular heterogeneity in non-transformed versus transformed cells. These features can be exploited for the development of therapies directed at specific cell subpopulations in precancerous lesions and within lung tumors.
Collapse
Affiliation(s)
- Sokviseth Moeng
- Center for Genomic and Precision Medicine, Texas A&M Institute of Biosciences and Technology, Texas Medical Center, Houston, TX 77030, USA; (S.M.); (A.D.C.-P.)
| | - Andres D. Chamorro-Parejo
- Center for Genomic and Precision Medicine, Texas A&M Institute of Biosciences and Technology, Texas Medical Center, Houston, TX 77030, USA; (S.M.); (A.D.C.-P.)
| | - Minsun S. Jeon
- Center for Epigenetics and Disease Prevention, Texas A&M Institute of Biosciences and Technology, Texas Medical Center, Houston, TX 77030, USA;
| | - James J. Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA;
| | - Kenneth S. Ramos
- Center for Genomic and Precision Medicine, Texas A&M Institute of Biosciences and Technology, Texas Medical Center, Houston, TX 77030, USA; (S.M.); (A.D.C.-P.)
| |
Collapse
|
283
|
Zhang Y. Enhancing rectal cancer liver metastasis prediction: Magnetic resonance imaging-based radiomics, bias mitigation, and regulatory considerations. World J Gastrointest Oncol 2025; 17:102151. [PMID: 39958549 PMCID: PMC11756008 DOI: 10.4251/wjgo.v17.i2.102151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/20/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025] Open
Abstract
In this article, we comment on the article by Long et al published in the recent issue of the World Journal of Gastrointestinal Oncology. Rectal cancer patients are at risk for developing metachronous liver metastasis (MLM), yet early prediction remains challenging due to variations in tumor heterogeneity and the limitations of traditional diagnostic methods. Therefore, there is an urgent need for non-invasive techniques to improve patient outcomes. Long et al's study introduces an innovative magnetic resonance imaging (MRI)-based radiomics model that integrates high-throughput imaging data with clinical variables to predict MLM. The study employed a 7:3 split to generate training and validation datasets. The MLM prediction model was constructed using the training set and subsequently validated on the validation set using area under the curve (AUC) and dollar-cost averaging metrics to assess performance, robustness, and generalizability. By employing advanced algorithms, the model provides a non-invasive solution to assess tumor heterogeneity for better metastasis prediction, enabling early intervention and personalized treatment planning. However, variations in MRI parameters, such as differences in scanning resolutions and protocols across facilities, patient heterogeneity (e.g., age, comorbidities), and external factors like carcinoembryonic antigen levels introduce biases. Additionally, confounding factors such as diagnostic staging methods and patient comorbidities require further validation and adjustment to ensure accuracy and generalizability. With evolving Food and Drug Administration regulations on machine learning models in healthcare, compliance and careful consideration of these regulatory requirements are essential to ensuring safe and effective implementation of this approach in clinical practice. In the future, clinicians may be able to utilize data-driven, patient-centric artificial intelligence (AI)-enhanced imaging tools integrated with clinical data, which would help improve early detection of MLM and optimize personalized treatment strategies. Combining radiomics, genomics, histological data, and demographic information can significantly enhance the accuracy and precision of predictive models.
Collapse
Affiliation(s)
- Yuwei Zhang
- Department of Digital Health, Northern Medical Center, Middletown, NY 10940, United States
| |
Collapse
|
284
|
Juza A, Kołodziej-Spirodek L, Gutkowski K, Partyka M, Dąbrowski M. Distinguishing exocrine pancreas disease-associated diabetes from type 2 diabetes based on anthropometric and metabolic parameters. World J Diabetes 2025; 16:95102. [PMID: 39959260 PMCID: PMC11718472 DOI: 10.4239/wjd.v16.i2.95102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/28/2024] [Accepted: 10/29/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Adult-onset diabetes is most often considered to be type 2 diabetes. However, other types of diabetes can develop in adults, including exocrine pancreas disease-associated diabetes, also called type 3c diabetes. Differential diagnosis between these types of diabetes still remains a diagnostic challenge. AIM To define anthropometric and laboratory markers that will allow for early diagnosis of pancreatic disease-associated diabetes. METHODS The study group included 44 patients with pancreatogenic diabetes (26 with pancreatic cancer and 18 with chronic pancreatitis), while the control group consisted of 35 patients with type 2 diabetes. We analyzed several parameters, including sex, age, body mass index (BMI), fasting plasma glucose, fasting C-peptide and insulin with homeostasis model assessment of insulin resistance (HOMA-IR) index calculation, adrenomedullin, adiponectin and creatinine levels with epidermal growth factor receptor (eGFR) calculation. We also developed an equation, termed type 3c diabetes index, which utilized BMI, fasting insulin and adrenomedullin levels, and eGFR to better identify patients with type 3c diabetes. RESULTS Compared to patients with type 2 diabetes, patients with pancreatogenic diabetes had significantly lower BMI (25.11 ± 4.87 kg/m2 vs 30.83 ± 5.21 kg/m2), fasting C-peptide (0.81 ± 0.42 nmol/L vs 1.71 ± 0.80 nmol/L), insulin (76.81 ± 63.34 pmol/L vs 233.19 ± 164.51 pmol/L) and HOMA-IR index, despite similar fasting plasma glucose levels. Patients with pancreatogenic diabetes also had lower adrenomedullin levels (0.41 ± 0.25 ng/mL vs 0.63 ± 0.38 ng/mL) but higher adiponectin levels (13.08 ± 7.20 μg/mL vs 8.28 ± 4.01 μg/mL) and eGFR levels (100.53 ± 21.60 mL/min/1.73 m2 vs 85.14 ± 19.24 mL/min/1.73 m2). Finally, patients with pancreatogenic diabetes had significantly lower Type 3c diabetes index values. CONCLUSION Patients with pancreatogenic diabetes differ from patients with type 2 diabetes in anthropometric and laboratory parameters. The type 3c diabetes index had the highest discriminating value, above any single parameter.
Collapse
Affiliation(s)
- Anna Juza
- College of Medical Sciences, Institute of Medical Sciences, University of Rzeszów, Rzeszów 35-959, Poland
- Diabetic Outpatient Clinic & Department of Gastroenterology and Hepatology with the Subunit of Internal Diseases, University Clinical Hospital, Rzeszów 35-055, Poland
| | - Lilianna Kołodziej-Spirodek
- Diabetic Outpatient Clinic & Department of Gastroenterology and Hepatology with the Subunit of Internal Diseases, University Clinical Hospital, Rzeszów 35-055, Poland
| | - Krzysztof Gutkowski
- College of Medical Sciences, Institute of Medical Sciences, University of Rzeszów, Rzeszów 35-959, Poland
| | - Mariusz Partyka
- College of Medical Sciences, Institute of Medical Sciences, University of Rzeszów, Rzeszów 35-959, Poland
- Department of Internal Diseases, Nephrology and Endocrinology with the Nuclear Medicine Laboratory and the Dialysis Center & Endocrinology Outpatient Clinic, Clinical Provincial Hospital No. 2 in Rzeszów, Rzeszów 35-301, Poland
| | - Mariusz Dąbrowski
- College of Medical Sciences, Institute of Medical Sciences, University of Rzeszów, Rzeszów 35-959, Poland
| |
Collapse
|
285
|
Ositelu KC, Peesay T, Garcia C, Akhter N. Life's Essential 8 and Cardiovascular Disease in Breast Cancer Survivors. Curr Cardiol Rep 2025; 27:55. [PMID: 39954113 DOI: 10.1007/s11886-025-02216-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
PURPOSE OF REVIEW To explore the role of optimal cardiovascular health as defined by the Life's Essential 8 in breast cancer survivors. RECENT FINDINGS Optimal cardiovascular health may be associated with decreased cancer mortality. Breast cancer survivors may derive additional benefit from obtaining ideal cardiovascular health as defined by the Life's Essential 8. Certain components of the Life's Essential 8 may impact cardiovascular risk but also cancer mortality, and risk for cancer therapy related cardiac dysfunction. Continued physical activity, avoidance of smoking, and control of lipids and blood pressure are beneficial in breast cancer survivors. More study is needed to define the role of anti-hyperglycemic agents, BMI, and sleep on CVD risk in breast cancer survivors. The Life's Essential 8 can be a tool to inform clinicians regarding a breast cancer survivor's disease risk and to identify potential areas of improvement.
Collapse
Affiliation(s)
- Kamari C Ositelu
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Tejasvi Peesay
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Carol Garcia
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA
| | - Nausheen Akhter
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 N. St. Clair, Suite 600, Chicago, IL, 60611, USA.
- Division of Cardiovascular Medicine, Northwestern University Feinberg School of Medicine, 676 N St. Clair, Suite 600, Chicago, IL, 60611, USA.
| |
Collapse
|
286
|
Alaca Y. Machine learning via DARTS-Optimized MobileViT models for pancreatic Cancer diagnosis with graph-based deep learning. BMC Med Inform Decis Mak 2025; 25:81. [PMID: 39955532 PMCID: PMC11830204 DOI: 10.1186/s12911-025-02923-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 02/07/2025] [Indexed: 02/17/2025] Open
Abstract
The diagnosis of pancreatic cancer presents a significant challenge due to the asymptomatic nature of the disease and the fact that it is frequently detected at an advanced stage. This study presents a novel approach combining graph-based data representation with DARTS-optimised MobileViT models, with the objective of enhancing diagnostic accuracy and reliability. The images of the pancreatic CT were transformed into graph structures using the Harris Corner Detection algorithm, which enables the capture of complex spatial relationships. Subsequently, the graph representations were processed using MobileViT models that had been optimised with Differentiable Architecture Search (DARTS), thereby enabling dynamic architectural adaptation. To further enhance classification accuracy, advanced machine learning algorithms, including K-Nearest Neighbours (KNN), Support Vector Machines (SVM), Random Forest (RF), and XGBoost, were applied. The MobileViTv2_150 and MobileViTv2_200 models demonstrated remarkable performance, with an accuracy of 97.33% and an F1 score of 96.25%, surpassing the capabilities of traditional CNN and Vision Transformer models. This innovative integration of graph-based deep learning and machine learning techniques demonstrates the potential of the proposed method to establish a new standard for early pancreatic cancer diagnosis. Furthermore, the study highlights the scalability of this approach for broader applications in medical imaging, which could lead to improved patient outcomes.
Collapse
Affiliation(s)
- Yusuf Alaca
- Department of Computer Engineering, Hitit University, Çorum, 19500, Turkey.
| |
Collapse
|
287
|
Chen H, Liu L, Xing G, Zhang D, A. N, Huang J, Li Y, Zhao G, Liu M. Exosome tropism and various pathways in lung cancer metastasis. Front Immunol 2025; 16:1517495. [PMID: 40028322 PMCID: PMC11868168 DOI: 10.3389/fimmu.2025.1517495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Lung cancer, characterized by its high morbidity and mortality rates, has the capability to metastasize to various organs, thereby amplifying its detrimental impact and fatality. The metastasis of lung cancer is a complex biological phenomenon involving numerous physiological transformations. Exosomes, small membranous vesicles enriched with biologically active components, are pivotal in mediating intercellular communication and regulating physiological functions due to their specificity and stability. Extensive research has elucidated the production and functions of exosomes in cancer contexts. Multitude of evidence demonstrates a strong association between lung cancer metastasis and exosomes. Additionally, the concept of the pre-metastatic niche is crucial in the metastatic process facilitated by exosomes. This review emphasizes the role of exosomes in mediating lung cancer metastasis and their impact on the disease's development and the progression to other tissues. Furthermore, it explores the potential of exosomes as biomarkers for lung cancer metastasis, offering significant insights for future clinical advancements.
Collapse
Affiliation(s)
- Hui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Lin Liu
- Department of Drug Dispensing, The Third Hospital of Mianyang, Sichuan Mental Health Center, MianYang, China
| | - Gang Xing
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Dan Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Niumuqie A.
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianlin Huang
- Department of Pharmacy, Luzhou Naxi District People’s Hospital, Luzhou, China
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ge Zhao
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Minghua Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
288
|
Xie Q, Cui M. The prognosis impact of NACT-IDS and PDS in advanced ovarian cancer: a systematic review and meta-analysis. J Gynecol Oncol 2025; 36:36.e61. [PMID: 40017161 DOI: 10.3802/jgo.2025.36.e61] [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: 12/07/2023] [Revised: 09/30/2024] [Accepted: 11/25/2024] [Indexed: 03/01/2025] Open
Abstract
We aimed to explore the prognostic implications of neoadjuvant chemotherapy and interval debulking surgery (NACT-IDS) compared to primary debulking surgery (PDS) in patients diagnosed with advanced ovarian cancer by meta-analysis. The search was conducted across PubMed, Web of Science, Cochrane, Wanfang Data, China National Knowledge Infrastructure, and the Chinese BioMedical Literature Database to identify pertinent studies examining the prognostic implications of NACT-IDS versus PDS in patients with advanced ovarian cancer. As of September 11, 2023, a total of 29 articles were ultimately included, encompassing 12,916 patients with advanced ovarian cancer in this meta-analysis. NACT-IDS groups exhibited a higher satisfactory tumor reduction rate (odds ratio [OR]=2.06; 95% confidence interval [CI]=1.53 to 2.76; p<0.001). NACT-IDS effectively reduced the risk of adverse cardiac events (OR=0.36; 95% CI=0.17 to 0.80; p=0.012), surgical site infections (OR=0.42; 95% CI=0.29 to 0.60; p<0.001), and embolic complications (OR=0.43; 95% CI=0.24 to 0.75; p=0.003) in patients with advanced ovarian cancer. Compared to NACT-IDS therapy for International Federation of Gynecology and Obstetrics (FIGO) III-IV ovarian cancer patients (OR=1.66; 95% CI=1.24 to 2.23; p=0.009), NACT-IDS groups exhibited a higher satisfactory tumor reduction rate for FIGO IIIC-IV (OR=2.35; 95% CI=1.50 to 3.70; p<0.001). NACT-IDS effectively enhances the satisfactory tumor reduction rate, especially for patients with stage IIIC and IV, and decreases postoperative complications among patients with advanced ovarian cancer.
Collapse
Affiliation(s)
- Qiuxian Xie
- Department of Gynaecology, Chaozhou general hospital, Chaozhou, China
| | - Min Cui
- Department of Obstetrics and Gynecology, Taiyuan Central Hospital, Taiyuan, China.
| |
Collapse
|
289
|
Duchon M, Naik R, Lecuru F, Ferron G, Cornou C, Madad Zadeh S, Pomel C. Management of Recurrence in Ovarian Cancer-The Role of Surgery and HIPEC with Relevance to BRCA Testing in a PARPi Landscape. Cancers (Basel) 2025; 17:646. [PMID: 40002241 PMCID: PMC11852647 DOI: 10.3390/cancers17040646] [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: 11/29/2024] [Revised: 01/22/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Background: The surgical and medical management of recurrent ovarian cancer is complex and requires a personalized approach based on several factors, including the timing of recurrence, the patient's performance status, previous treatment regimens, and the tumor's histology and molecular characteristics. Objectives: Five randomized trials-GOG-0213, DESKTOP III, SOC 1, HORSE, and CHIPOR-have been conducted and shed light on our practice. Results: Both the DESKTOP III and the SOC 1 trials support the benefit of secondary surgery. The GOG-0213 trial, however, did not show an overall survival benefit, confirming that surgery should not be offered to all patients with platinum-sensitive recurrent ovarian cancer and highlighting the importance of strict patient selection using evidence-based selection criteria including the AGO or iMODEL scores. In patients with a negative score, there may be a place for cytoreductive surgery plus HIPEC in BRCA-negative cases following a course of chemotherapy, although current evidence shows no additional benefit for HIPEC when cytoreductive surgery is performed as an adjuvant procedure. Conclusions: Secondary surgery is recommended for platinum-sensitive patients when respecting AGO or iModel criteria. In addition, surgery plus HIPEC can be considered in BRCA-negative patients with an initial negative AGO or iMODEL score who show sufficient response following a course of neoadjuvant chemotherapy to then be considered operable.
Collapse
Affiliation(s)
- Mathilde Duchon
- Department of Surgical Oncology, Centre Jean-Perrin, 58, rue Montalembert, 63011 Clermont-Ferrand, France; (M.D.); (C.C.); (S.M.Z.)
| | - Raj Naik
- British Surgical Gynaecological Oncology Group (BSGOG), Newcastle upon Tyne NE2 4DJ, UK;
| | - Fabrice Lecuru
- Department of Surgical Oncology, Institut Curie, 26 Rue d’Ulm, 75005 Paris, France;
| | - Gwenaël Ferron
- Department of Surgical Oncology, IUCT Oncopole, Institut Universitaire du Cancer de Toulouse, 31100 Toulouse, France;
| | - Caroline Cornou
- Department of Surgical Oncology, Centre Jean-Perrin, 58, rue Montalembert, 63011 Clermont-Ferrand, France; (M.D.); (C.C.); (S.M.Z.)
| | - Sabrina Madad Zadeh
- Department of Surgical Oncology, Centre Jean-Perrin, 58, rue Montalembert, 63011 Clermont-Ferrand, France; (M.D.); (C.C.); (S.M.Z.)
| | - Christophe Pomel
- Department of Surgical Oncology, Centre Jean-Perrin, 58, rue Montalembert, 63011 Clermont-Ferrand, France; (M.D.); (C.C.); (S.M.Z.)
| |
Collapse
|
290
|
Chen J, Xiang Q, Zheng XJ, Jiang XY. Predictive model for postoperative pneumonia in patients with esophageal cancer after esophagectomy. Front Oncol 2025; 15:1529308. [PMID: 40027123 PMCID: PMC11868259 DOI: 10.3389/fonc.2025.1529308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/31/2025] [Indexed: 03/05/2025] Open
Abstract
Background Pneumonia is one of the most common complications after esophagectomy and a risk factor affecting postoperative survival of esophageal cancer. The aim of this study was to identify risk factors and construct a predictive model for postoperative pneumonia (POP) in esophageal cancer. Methods This retrospective cohort study included esophageal cancer patients who underwent therapeutic esophagectomy from June 2019 to December 2023. Least absolute shrinkage and selection operator (LASSO) regression was used to screen predictive factors for POP, and a nomogram was constructed based on the selected predictive factors after screening. The performance of the model was evaluated using the area under the receiver operating characteristic curve (AUC), calibration curve, and decision curve analysis (DCA). Results A total of 667 esophageal cancer patients who underwent esophagectomy were included, of whom 61 (9.1%) developed postoperative pneumonia. After LASSO regression analysis, factors independently associated with POP included mechanical ventilation for more than 2 days (P=0.000) and blood transfusion (P=0.003). A nomogram was constructed based on these independent risk factors. The AUC of the predictive model for POP was 0.839 (95%CI: 0.768-0.911). The internal verification result showed a good discriminative power and the DCA results demonstrated a good predictive value. Conclusion The predictive model constructed in this study can predict the risk of POP in patients with esophageal cancer, and may promote early intervention for high-risk patients by clinicians to reduce the incidence of POP.
Collapse
Affiliation(s)
| | | | | | - Xiao-yan Jiang
- Department of Healthcare-associated Infection Control Center, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
291
|
Wang Y, Guo E, Zou M, Lv C, Cui Y, Zhai S, Sang S, Xiong K, Yang X, Zhuang S, Gu Y, Liang H. Unraveling immune heterogeneity across pan-cancer and deep insights in lung adenocarcinoma based on alternative splicing. J Leukoc Biol 2025; 117:qiae104. [PMID: 38758950 DOI: 10.1093/jleuko/qiae104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/28/2024] [Accepted: 04/16/2024] [Indexed: 05/19/2024] Open
Abstract
Alternative splicing (AS) participates in tumor development and tumor microenvironment formation. However, the landscape of immune-infiltrating AS events in pan-cancer and mechanisms of AS in lung adenocarcinoma (LUAD) have not been comprehensively characterized. We systematically profiled the immune-infiltrating AS event landscape of pan-cancer using data from The Cancer Genome Atlas, analyzing both commonalities and specific characteristics among different cancer types. We found that AS events tend to occur specifically in one cancer type rather than in multiple cancer types. AS events were used to classify 512 LUAD samples into 2 subtypes by unsupervised clustering: the aberrant splicing subtype and the immune-infiltrating subtype. The 2 subtypes showed significant differences in clinicopathology, prognosis, transcriptomics, genomics, and immune microenvironment. We constructed a classification signature comprising 10 genes involved in 14 AS events using logistic regression. The robustness of the signature was validated in 3 independent datasets using survival analysis. To explore AS mechanisms in LUAD, we constructed subtype-specific coexpression networks using Pearson correlation analysis. AS event of AKT3 regulated by splicing factor ENOX1 was associated with poor prognosis in LUAD. Overall, we outline AS events associated with immune infiltration in pan-cancer, and this study provides insights into AS mechanisms in LUAD patient classification.
Collapse
Affiliation(s)
- Yuquan Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Laboratorary-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Erliang Guo
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Min Zou
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, No. 157 Baojian Road, NanGang District, Harbin, Heilongjiang Province, 150081, China
| | - Chen Lv
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, No. 157 Baojian Road, NanGang District, Harbin, Heilongjiang Province, 150081, China
| | - Yanrui Cui
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, No. 157 Baojian Road, NanGang District, Harbin, Heilongjiang Province, 150081, China
| | - Songmei Zhai
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, No. 157 Baojian Road, NanGang District, Harbin, Heilongjiang Province, 150081, China
| | - Shaocong Sang
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, No. 157 Baojian Road, NanGang District, Harbin, Heilongjiang Province, 150081, China
| | - Kai Xiong
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, No. 157 Baojian Road, NanGang District, Harbin, Heilongjiang Province, 150081, China
| | - Xiuqi Yang
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, No. 157 Baojian Road, NanGang District, Harbin, Heilongjiang Province, 150081, China
| | - Shuping Zhuang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Laboratorary-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Yunyan Gu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, No. 157 Baojian Road, NanGang District, Harbin, Heilongjiang Province, 150081, China
| | - Haihai Liang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Laboratorary-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, No. 157 Baojian Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| |
Collapse
|
292
|
Mandal S, Jana P, Naskar R, Halder A, Bera B, Chattopadhyay K, Mondal TK. An Investigation into Substitution-Kinetics, Biomolecular Responses and Multimodal Anticancer Potential of a Dihalide Pd(II) Complex. Chem Asian J 2025:e202401832. [PMID: 39945662 DOI: 10.1002/asia.202401832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/08/2025] [Accepted: 02/13/2025] [Indexed: 03/05/2025]
Abstract
This study addresses a novel palladium dihalide complex, cis-[Pd(PCAH)Cl₂] (C1), as a promising anticancer agent. XRD analysis reveals a deformed square planar geometry stabilized by hydrogen bonds and π•••π interactions. The M-Cl bonds in C1 demonstrate susceptibility to nucleophilic substitution by 2,2'-bipyridine (Bpy), with kinetic parameters evaluated using spectrophotometry. Fluorometric and spectrophotometric investigations demonstrate that C1 binds to CT DNA and protein with an avidity of around 105 M-1. The interaction with DNA is multifaceted, employing covalent bonding and intercalation, as supported by viscosity measurements. Fluorescence lifetime experiments illustrate that C1 produces static dampening of BSA fluorescence, implying structural adjustments near the tryptophan residue, further corroborated by spectroscopic analyses. The pair's (BSA and C1) FRET distance has also been computed. In vitro cytotoxicity tests suggest that C1 selectively suppresses the growth of breast carcinoma, MDA-MB-231 with IC50=20±2.64 μM, while showing minimal effects on non-cancerous HEK-293 cells. The mechanism of action includes the creation of ROS, leading to mitochondrial apoptosis, as evidenced by various assays, including annexin-V-FITC/PI labeling. Overall, complex C1 exhibits encouraging promise as a selective anticancer drug with a ROS-triggered apoptotic mechanism, particularly effective against breast carcinoma MDA-MB-231 cells.
Collapse
Affiliation(s)
- Subrata Mandal
- Department of Chemistry, Jadavpur University, Jadavpur, Kolkata, 700032, India
| | - Pulak Jana
- Structural Biology & Bio-Informatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mallick Road, Kolkata, 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Rahul Naskar
- Department of Chemistry, Jadavpur University, Jadavpur, Kolkata, 700032, India
| | - Arpan Halder
- Department of Chemistry, Jadavpur University, Jadavpur, Kolkata, 700032, India
| | - Biswajit Bera
- Department of Chemistry, Jadavpur University, Jadavpur, Kolkata, 700032, India
| | - Krishnananda Chattopadhyay
- Structural Biology & Bio-Informatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mallick Road, Kolkata, 700032, India
| | - Tapan K Mondal
- Department of Chemistry, Jadavpur University, Jadavpur, Kolkata, 700032, India
| |
Collapse
|
293
|
Chen X, Zhang F, Lu C, Wu R, Yang B, Liao T, Du B, Wu F, Ding J, Fang S, Zhao Z, Chen M, Shu G, Chen W, Ji J. Lactate-Fueled Theranostic Nanoplatforms for Enhanced MRI-Guided Ferroptosis Synergistic with Immunotherapy of Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2025; 17:9155-9172. [PMID: 39901437 DOI: 10.1021/acsami.4c21890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Treatment for hepatocellular carcinoma (HCC) may be improved with ferroptosis, a regulated form of cell death. However, the sensitivity of HCC to ferroptosis was strongly limited by lactic acid. In this study, a platelet membrane (PM)-engineered nanoparticle loaded with erastin, superparamagnetic iron oxide nanoparticles (SPIO) and lactate oxidase (LOX) (termed PM@ESL NPs) was designed for magnetic resonance imaging (MRI)-guided enhanced ferroptosis-immunotherapy of HCC. It was found that PM@ESL NPs could actively accumulate into the tumor due to the tumor-homing ability of PM. Subsequently, PM@ESL NPs could effectively enhance the sensitivity of HCC to ferroptosis by removing the lactic acid in the tumor. The removal of lactic acid also produces hydrogen peroxide (H2O2), which therefore converted into the cytotoxic hydroxyl radicals by the reaction of H2O2 with Fe2+/Fe3+ released from SPIO. Due to the combined ferroptosis and chemodynamic therapy (CDT), PM@ESL NPS showed a strong ability to induce immunogenic cell death (ICD), which could effectively suppress the growth and metastasis of HCC when combined with αPD-L1 immunotherapy. Furthermore, the incorporation of SPIO endows PM@ESL NPs with an outstanding MRI-T2 monitoring capability for HCC treatment. In conclusion, this study introduces a pioneering MRI-guided approach that enhances ferroptosis in tumors and synergistically improves immunotherapy.
Collapse
Affiliation(s)
| | - Feng Zhang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Chenying Lu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Ronghua Wu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medcine, Lishui University, Lishui 323000, China
| | - Baozhu Yang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Tingting Liao
- College of pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 310015, China
| | - Baojie Du
- Shanxi Bethune Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Taiyuan 030032, China
| | - Fazong Wu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Jiayi Ding
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Shiji Fang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Zhongwei Zhao
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medcine, Lishui University, Lishui 323000, China
| | - Minjiang Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Gaofeng Shu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Weiqian Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Jiansong Ji
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- College of pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 310015, China
- Clinical College of The Affiliated Central Hospital, School of Medcine, Lishui University, Lishui 323000, China
| |
Collapse
|
294
|
Deng Z, Jin X, Liu B, Zhen H, Wang X. Unveiling the prognostic significance of RNA editing-related genes in colon cancer: evidence from bioinformatics and experiment. Eur J Med Res 2025; 30:94. [PMID: 39940052 PMCID: PMC11823094 DOI: 10.1186/s40001-025-02335-7] [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: 10/19/2024] [Accepted: 01/26/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND RNA editing is recognized as a crucial factor in cancer biology. Its potential application in predicting the prognosis of colon adenocarcinoma (COAD) remains unexplored. METHODS RNA editing data of COAD patients were downloaded from the Synapse database. LASSO regression was used to construct the risk model and verified by the receiver operating characteristic (ROC) curve. GO and KEGG enrichment analyses were performed to delineate the biological significance of the differentially expressed genes. Finally, differential analysis and immunohistochemistry were used to verify the expression of adenosine deaminase 1 (ADAR1). RESULTS We evaluated a total of 4079 RNA editing sites in 514 COAD patients from Synapse database. A prognostic signature was constructed based on five genes were significantly associated with the prognosis of COAD patients including GNL3L, NUP43, MAGT1, EMP2, and ARSD. Univariate and multivariate Cox regression analysis revealed that RNA editing-related genes (RERGs)-related signature was an independent risk factor for COAD. Moreover, Experimental evidence shows that ADAR1 is highly expressed in colon adenocarcinoma and silencing ADAR1 can inhibit cancer cell proliferation. CONCLUSION We established a prognostic model based on five RERGs with strong predictive value. This model not only serves as a foundation for a novel prognostic tool but also facilitates the identification of potential drug candidates for treating COAD.
Collapse
Affiliation(s)
- Zhengcong Deng
- Hubei Third People's Hospital, Wuhan, 430033, Hubei, China
- Wuhan Donghu New Technology Development Zone Disease Prevention and Control Center, Wuhan, 430200, Hubei, China
| | - Xueqin Jin
- Hubei Third People's Hospital, Wuhan, 430033, Hubei, China
| | - Bingxue Liu
- Medical School, Jianghan University, Wuhan, 430056, Hubei, China
| | - Hongyan Zhen
- Medical School, Jianghan University, Wuhan, 430056, Hubei, China
| | - Xiang Wang
- Medical School, Jianghan University, Wuhan, 430056, Hubei, China.
- Wuhan University of Arts and Science, Wuhan, 430345, Hubei, China.
| |
Collapse
|
295
|
Sowndarya A, Daniel Thangadurai T, Thomas NG, Sreedharan R, Anil S, Manjubaashini N, Satheesh Babu TG, Megha Kumar S. Effect of surface-engineered AuNPs on gene expression, bacterial interaction, protein denaturation, and toxicology assay: an in vitro and in vivo model. J Mater Chem B 2025; 13:2409-2417. [PMID: 39812144 DOI: 10.1039/d4tb01731e] [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: 01/16/2025]
Abstract
We investigated the in vitro and in vivo uses of pamoic acid functionalized gold nanoparticles (PA@AuNPs), with a focus on determining their safety and potential toxicity in living beings. To test this theory, the bacterial interaction of PA@AuNPs was studied using Escherichia coli, Staphylococcus aureus, and Pseudomonas aeruginosa cultures, as well as the inhibition of the bovine serum albumin (BSA) protein. The real-time polymerase chain reaction (RT-PCR) is used to measure the expression of target genes. PA@AuNPs caused dose-dependent cell death in MDA-MB-231, a triple-negative breast cancer (BC) cell line, with an LC50 of -42.23 μL mL-1. It also caused apoptosis in BC cells. The results indicated that in the early weeks, inflammatory cells (mostly neutrophils and macrophages) penetrated the connective tissue, but in the latter weeks, a substantial number of fibroblasts and fibrocytes were identified. Changes in vascular channels, extravasated red blood cells (RBCs), and necrosis are all indicators of growing tissue pathology. These data could point to a dynamic process including an anti-inflammatory response followed by tissue remodeling or repair. These findings show that PA@AuNPs were not hazardous to the tested Sprague Dawley rats, are highly biocompatible, and can be used in a variety of biological applications.
Collapse
Affiliation(s)
- A Sowndarya
- Department of Chemistry and Centre for Research and Development, KPR Institute of Engineering and Technology, Coimbatore 641407, Tamilnadu, India.
| | - T Daniel Thangadurai
- Department of Chemistry and Centre for Research and Development, KPR Institute of Engineering and Technology, Coimbatore 641407, Tamilnadu, India.
| | - Nebu George Thomas
- Pushpagiri Institute of Medical Sciences and Research Centre, Thiruvalla 689101, Kerala, India
- Pushpagiri College of Dental Sciences, Thiruvalla 689101, Kerala, India
| | - Renjith Sreedharan
- Jubilee Mission Medical College and Research Institute, Thrissur 680005, Kerala, India
| | - Sukumaran Anil
- Pushpagiri Institute of Medical Sciences and Research Centre, Thiruvalla 689101, Kerala, India
- Oral Health Institute, Department of Dentistry, Hamad Medical Corporation Doha, Qatar
| | - N Manjubaashini
- National Centre for Nanoscience and Nanotechnology, University of Madras, Chennai 600025, India
| | - T G Satheesh Babu
- Department of Chemistry, Amrita School of Physical Sciences Coimbatore, Amrita Vishwa Vidyapeetham, India
- Amrita Biosensor Research Lab, Amrita School of Engineering Coimbatore, Amrita Vishwa Vidyapeetham, India
| | - S Megha Kumar
- Department of Chemistry, Amrita School of Physical Sciences Coimbatore, Amrita Vishwa Vidyapeetham, India
- Amrita Biosensor Research Lab, Amrita School of Engineering Coimbatore, Amrita Vishwa Vidyapeetham, India
| |
Collapse
|
296
|
Liu Y, Shen Y, Luo P, Wu S, Wang Y, Deng J, Deng L, Wang F, Jin J, Jiang J. Identification of HES4 as a novel prognostic marker and therapeutic target in hepatocellular carcinoma. Discov Oncol 2025; 16:156. [PMID: 39934570 DOI: 10.1007/s12672-025-01915-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
Hairy and enhancer of Split 4 (HES4) is thought to have a substantial impact on the pathogenesis and progression of malignancies. However, the prognostic significance and mechanism of HES4 have not been reported in Hepatocellular carcinoma (HCC). A comprehensive bioinformatics analysis of HES4 expression, clinicopathological characteristics, tumor microenvironment status, and drug sensitivity were performed based on TCGA, GTEx, and GEO. Paired HCC samples and cell lines were used to validate the dysfunction of HES4 in vitro. The expression of HES4 at both mRNA and protein levels was significantly upregulated in HCC tissues. High level of HES4 was associated with unfavorable outcomes. Enrichment analysis demonstrated strong associations of HES4 with HCC progression pathways. In addition, elevated HES4 expression was positively correlated with increased sensitivity to various chemotherapy drugs and associated with resistance to immunotherapy. As a transcription factor, the target genes regulated by HES4 were mostly risky genes, and a novel prediction model based on HES4 target genes was generated for HCC risk stratification. The AUCs of 1-, 3-, and 5-year year overall survival (OS) were 0.829, 0.732, and 0.700, respectively. HES4 overexpression is associated with poor clinical outcomes and tumor progression. HES4 may serve as a novel prognostic marker and therapeutic target in HCC.
Collapse
Affiliation(s)
- Yungang Liu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Department of Oncology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Ying Shen
- Department of Oncology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Peipei Luo
- Department of Gastroenterology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China
- Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Yue Wang
- Department of Oncology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Jianzhong Deng
- Department of Oncology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Linghui Deng
- Department of Oncology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Fang Wang
- Department of Oncology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Jianhua Jin
- Department of Oncology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China.
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China.
- Institute of Cell Therapy, Soochow University, Changzhou, China.
| |
Collapse
|
297
|
Roboubi A, Wasielewski E, Bordier S, Turlotte A, Pavaut G, Scherpereel A, Cortot A, Gauvain C. Impact of corticosteroids on the efficacy of first-line pembrolizumab plus chemotherapy in patients with advanced non-small-cell lung cancer. Ther Adv Med Oncol 2025; 17:17588359251318160. [PMID: 39935765 PMCID: PMC11811968 DOI: 10.1177/17588359251318160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Background Systemic corticosteroids (SCs) are associated with reduced survival in patients with advanced non-small-cell lung cancer (NSCLC) receiving immune checkpoint inhibitor (ICI) monotherapy. However, the current first-line standard of care usually involves combined chemotherapy (CT) and ICIs, and the effect of SCs on survival under combined CT and ICI has never been studied. Objectives To investigate the association between SC therapy and survival under CT-ICI in advanced-stage NSCLC patients. Design We performed a multicenter retrospective cohort study of all advanced-stage NSCLC patients receiving first-line CT-ICI. Methods The primary endpoint was progression-free survival (PFS) according to SC exposure status (⩾10 mg/day), adjusted in a multivariate Cox model for the following confounders: age, performance status, hospital admission prior to treatment, number of metastatic sites, brain metastases, bone metastases, PD-L1 status, and histological subtype. Multivariate analyses also explored the association between dosage and SC exposure duration and PFS. Results Of the 193 included patients, 43 (22.3%) were receiving SCs, mainly because of symptomatic brain metastases (in 25/43 cases, 58%). In multivariate analysis, SC therapy at a 10 mg/day threshold was not associated with PFS (hazard ratio (HR) = 1.25, 95% confidence interval (CI) 0.77-2.03, p = 0.35). However, SC dose was negatively associated with PFS (HR = 1.08 per 10 mg/day increment, 95% CI 1.01-1.16, p = 0.01) especially at doses ⩾60 mg/day (HR = 3.27 per 10 mg/day increment, 95% CI 2.01-5.35, p < 0.001). Duration of SC therapy was not associated with PFS (HR = 0.97, 95% CI 0.81-1.15, p = 0.71), but SC therapy ⩾4 weeks prior to CT-ICI was associated with shorter PFS (HR = 1.07, 95% CI: 1.01-1.14, p = 0.028). Conclusion In this group of patients receiving first-line CT-ICI for advanced NSCLC, SCs at ⩾60 mg/day were associated with shorter PFS, but lower doses were not. Prolonged SC therapy prior to CT-ICI was associated with shorter PFS. Larger studies are required to confirm these results.
Collapse
Affiliation(s)
- Amytis Roboubi
- Service de Pneumologie et Oncologie Thoracique, Institut Cœur Poumon, CHU Lille, Lille, France
| | - Eric Wasielewski
- Service de Pneumologie et Oncologie Thoracique, Institut Cœur Poumon, CHU Lille, Lille, France
| | | | | | - Geoffrey Pavaut
- Service de Pneumologie, Clinique Teissier, Valenciennes, France
| | - Arnaud Scherpereel
- Service de Pneumologie et Oncologie Thoracique, Institut Cœur Poumon, CHU Lille, Lille, France
- Univ. Lille, CHU Lille, INSERM U1189, OncoThAI, Lille, France
| | - Alexis Cortot
- Service de Pneumologie et Oncologie Thoracique, Institut Cœur Poumon, CHU Lille, Lille, France
- Univ. Lille, CHU Lille, CNRS, Inserm, Institut Pasteur de Lille, UMR9020—UMR-S 1277, Canther, France
| | - Clément Gauvain
- Service de Pneumologie et Oncologie Thoracique, Institut Cœur Poumon, CHU de Lille, Boulevard du Pr Jules Leclercq, Lille 59000, France
| |
Collapse
|
298
|
Deng M, Wang P, Zhai Z, Liu Y, Cheng D, He L, Li S. A Triple-Responsive and Dual-NIR Emissive Fluorescence Probe for Precise Cancer Imaging and Therapy by Activating Pyroptosis Pathway. Anal Chem 2025; 97:2998-3008. [PMID: 39888040 DOI: 10.1021/acs.analchem.4c06015] [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: 02/01/2025]
Abstract
Revealing changes in the tumor microenvironment is crucial for understanding cancer and developing sensitive methods for precise cancer imaging and diagnosis. Intracellular hydrogen peroxide (H2O2) and microenvironmental factors (e.g., viscosity and polarity) are closely linked to various physiological and pathological processes, making them potential biomarkers for cancer. However, a triple-response theranostic probe for precise tumor imaging and therapy has not yet been achieved due to the lack of effective tools. Herein, we present a mitochondria-targeting near-infrared (NIR) fluorescent probe, VPH-5DF, capable of simultaneously monitoring H2O2, viscosity, and polarity through dual NIR channels. The probe specifically detects H2O2 via NIR emission (λem = 650 nm) and shows high sensitivity to microenvironmental viscosity/polarity in the deep NIR channel (λem ≈ 750 nm). Furthermore, the probe not only monitors mitochondrial polarity, viscosity, and fluctuations in endogenous/exogenous H2O2 levels but also distinguishes cancer cells from normal cells through multiple parameters. Additionally, VPH-5DF can be employed to monitor alterations in H2O2 levels, as well as changes in viscosity and polarity, during drug-induced pyroptosis in living cells. After treatment with VPH-5DF, chemotherapy-induced oxidative damage to the mitochondria in tumor cells activated the pyroptosis pathway, leading to a robust antitumor response, as evidenced in xenograft tumor models. Thus, this triple-response theranostic prodrug offers a new platform for precise in vivo cancer diagnosis and anticancer chemotherapy.
Collapse
Affiliation(s)
- Min Deng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang 421002, China
| | - Peipei Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang 421002, China
| | - Zibo Zhai
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang 421002, China
| | - Ying Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang 421002, China
| | - Dan Cheng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang 421002, China
- Department of Gastroenterology, Clinical Research Institute, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang 421002, PR China
| | - Longwei He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang 421002, China
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, PR China
| | - Songjiao Li
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang 421002, China
| |
Collapse
|
299
|
Ji Y, Zhang H, Gong FL, Liang JL, Wang SF, Sang YH, Zheng MF. The expression and functional role of proline-rich 15 in non-small cell lung cancer. Cell Death Dis 2025; 16:83. [PMID: 39929816 PMCID: PMC11811231 DOI: 10.1038/s41419-025-07373-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 01/05/2025] [Accepted: 01/21/2025] [Indexed: 02/13/2025]
Abstract
Proline-rich 15 (PRR15) is a protein primarily known for its role in placental development. This study investigates the expression, functional significance, and underlying mechanisms of PRR15 in non-small cell lung cancer (NSCLC). Our findings demonstrate significantly elevated PRR15 expression in NSCLC tissues compared to normal lung parenchyma, with higher expression correlating with adverse clinical outcomes. Single-cell RNA sequencing confirmed PRR15 overexpression within the malignant tumor cell population. PRR15 expression was elevated in NSCLC tissues from locally treated patients and in a panel of primary and established NSCLC cells. PRR15 depletion using shRNA or CRISPR/Cas9-mediated knockout significantly suppressed proliferation and migration, while promoting apoptosis in various NSCLC cells. Conversely, ectopic PRR15 overexpression using a lentiviral construct enhanced cell proliferation and migration. Mechanistic investigations implicated PRR15 in the activation of the Akt-mTOR signaling pathway. Inhibition of PRR15 expression via shRNA or CRISPR/Cas9-mediated knockout resulted in decreased Akt and S6K phosphorylation, while PRR15 overexpression led to augmented Akt-S6K signaling in primary human NSCLC cells. In vivo studies using xenograft models further validated the oncogenic role of PRR15, demonstrating that PRR15 knockdown suppressed tumor growth and attenuated Akt-mTOR activation. These findings collectively highlight the potential of PRR15 as a novel oncogenic driver and therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Yong Ji
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Han Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Fei-Long Gong
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Jia-Long Liang
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Sheng-Fei Wang
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Yong-Hua Sang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ming-Feng Zheng
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| |
Collapse
|
300
|
Hagemann AR, Hagemann IS, Mutch DG, Devor EJ, Malmrose PK, Zhang Y, Morrison AM, Thiel KW, Leslie KK. Enhancing Progestin Therapy with a Glucagon-Like Peptide 1 Agonist for the Conservative Management of Endometrial Cancer. Cancers (Basel) 2025; 17:598. [PMID: 40002193 PMCID: PMC11853405 DOI: 10.3390/cancers17040598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/24/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
OBJECTIVE Obesity is a major risk factor for endometrial cancer. In addition to hormone therapy with progestins, glucagon like peptide-1 receptor (GLP-1R) agonists such as semaglutide may be helpful to achieve weight loss during conservative treatment of endometrial hyperplasia or cancer. METHODS We theorized that the combination of semaglutide and the progestin levonorgestrel would be useful as a novel treatment or prevention regimen and tested this hypothesis using endometrial cancer cell lines and patient-derived organoids (PDOs). RESULTS Hec50, KLE, and Ishikawa endometrial cancer cells express GLP-1R, as determined by both qPCR and Western blotting, and GLP-1R agonist treatment induces GLP-1R mRNA transcription through positive feedback mechanisms in cell models. PDOs from six individuals with grade 1 endometrial carcinomas were treated with progesterone, levonorgestrel, semaglutide, or levonorgestrel + semaglutide. Multiple models demonstrated a significant reduction in viability in response to combinatorial treatment, and the effect was noted in models from both PR high- and PR low-expressing tumors. Most interesting was the induction not only of the membrane GLP-1R with treatment, but also the significant upregulation of nuclear and membrane progesterone receptors-PR and PGRMC1/2, respectively-indicating a potential positive feedback loop between semaglutide and progestins such as levonorgestrel. CONCLUSION In summary, we identify synergistic molecular cross-talk between the GLP-1R and steroid hormone receptor pathways, with the potential to enhance the anticancer activity of levonorgestrel when combined with semaglutide.
Collapse
Affiliation(s)
- Andrea R. Hagemann
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, and Alvin J. Siteman Cancer Center, St. Louis, MO 63110, USA; (A.R.H.); (I.S.H.); (D.G.M.)
| | - Ian S. Hagemann
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, and Alvin J. Siteman Cancer Center, St. Louis, MO 63110, USA; (A.R.H.); (I.S.H.); (D.G.M.)
- Department of Pathology and Immunology, Washington University, St. Louis, MO 63110, USA
| | - David G. Mutch
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, and Alvin J. Siteman Cancer Center, St. Louis, MO 63110, USA; (A.R.H.); (I.S.H.); (D.G.M.)
| | - Eric J. Devor
- Department of Obstetrics and Gynecology and the Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (E.J.D.); (Y.Z.); (A.M.M.)
| | - Paige K. Malmrose
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Yuping Zhang
- Department of Obstetrics and Gynecology and the Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (E.J.D.); (Y.Z.); (A.M.M.)
| | - Abigail M. Morrison
- Department of Obstetrics and Gynecology and the Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (E.J.D.); (Y.Z.); (A.M.M.)
| | - Kristina W. Thiel
- Department of Obstetrics and Gynecology and the Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (E.J.D.); (Y.Z.); (A.M.M.)
| | - Kimberly K. Leslie
- Department of Internal Medicine and the University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| |
Collapse
|