1
|
Xue N, Wen X, Wang Q, Shen Y, Qu Y, Xu Q, Chen S, Chen J. Establishing and validating models integrated with hematological biomarkers and clinical characteristics for the prognosis of non-esophageal squamous cell carcinoma patients. Ann Med 2025; 57:2483985. [PMID: 40152751 PMCID: PMC11956093 DOI: 10.1080/07853890.2025.2483985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/11/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND This study aimed to construct a novel model and validate its predictive power in non-esophageal squamous cell carcinoma (NESCC) patients. METHODS This retrospective study included 151 patients between October 2006 and September 2016. The LASSO Cox and Random Survival Forest (RSF) models were developed with the help of hematological biomarkers and clinical characteristics. The concordance index (C-index) was used to assess the prognostic power of the LASSO Cox model, RSF model, and TNM staging. Based on the risk scores of the LASSO Cox and RSF models, we divided patients into low-risk and high-risk subgroups. RESULTS We constructed two models in NESCC patients according to LASSO Cox regression and RSF models. The RSF model reached a C-index of 0.841 (95% CI: 0.792-0.889) in the primary cohort and 0.880 (95% CI: 0.830-0.930) in the validation cohort, which was higher than the C-index of the LASSO Cox model 0.656 (95% CI: 0.580-0.732) and 0.632 (95% CI: 0.542-0.720) in the two cohorts. The integrated C/D area under the ROC curve (AUC) values for the LASSO Cox and RSF models were 0.701 and 0.861, respectively. In both two models, Kaplan-Meier survival analysis and the estimated restricted mean survival time (RMST) values indicated that the low-risk subgroup had a better prognostic outcome than the high-risk subgroup (p < 0.05). CONCLUSIONS The RSF model has better prediction power than the LASSO Cox and the TNM staging models. It has a guiding value for the choice of individualized treatment in patients with NESCC.
Collapse
Affiliation(s)
- Ning Xue
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| | - Xiaoyan Wen
- Central Sterilization Supply Department, The Guanghua Stomatological College of Sun Yat-sen University, Hospital of Stomatology, SunYat-sen University, Guangzhou, P. R. China
| | - Qian Wang
- Department of radiation oncology, China–Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Yong Shen
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| | - Yuanye Qu
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| | - Qingxia Xu
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| | - Shulin Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Research Center for Translational Medicine, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, P. R. China
| | - Jing Chen
- Department of Clinical Laboratory, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, P. R. China
| |
Collapse
|
2
|
Chen Y, Zhou C, Zhang X, Chen M, Wang M, Zhang L, Chen Y, Huang L, Sun J, Wang D, Chen Y. Construction of a novel radioresistance-related signature for prediction of prognosis, immune microenvironment and anti-tumour drug sensitivity in non-small cell lung cancer. Ann Med 2025; 57:2447930. [PMID: 39797413 PMCID: PMC11727174 DOI: 10.1080/07853890.2024.2447930] [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: 06/30/2024] [Revised: 11/26/2024] [Accepted: 12/12/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is a fatal disease, and radioresistance is an important factor leading to treatment failure and disease progression. The objective of this research was to detect radioresistance-related genes (RRRGs) with prognostic value in NSCLC. METHODS The weighted gene coexpression network analysis (WGCNA) and differentially expressed genes (DEGs) analysis were performed to identify RRRGs using expression profiles from TCGA and GEO databases. The least absolute shrinkage and selection operator (LASSO) regression and random survival forest (RSF) were used to screen for prognostically relevant RRRGs. Multivariate Cox regression was used to construct a risk score model. Then, Immune landscape and drug sensitivity were evaluated. The biological functions exerted by the key gene LBH were verified by in vitro experiments. RESULTS Ninety-nine RRRGs were screened by intersecting the results of DEGs and WGCNA, then 11 hub RRRGs associated with survival were identified using machine learning algorithms (LASSO and RSF). Subsequently, an eight-gene (APOBEC3B, DOCK4, IER5L, LBH, LY6K, RERG, RMDN2 and TSPAN2) risk score model was established and demonstrated to be an independent prognostic factor in NSCLC on the basis of Cox regression analysis. The immune landscape and sensitivity to anti-tumour drugs showed significant disparities between patients categorized into different risk score subgroups. In vitro experiments indicated that overexpression of LBH enhanced the radiosensitivity of A549 cells, and knockdown LBH reversed the cytotoxicity induced by X-rays. CONCLUSION Our study developed an eight-gene risk score model with potential clinical value that can be adopted for choice of drug treatment and prognostic prediction. Its clinical routine use may assist clinicians in selecting more rational practices for individuals, which is important for improving the prognosis of NSCLC patients. These findings also provide references for the development of potential therapeutic targets.
Collapse
Affiliation(s)
- Yanliang Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Chan Zhou
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiaoqiao Zhang
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Min Chen
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Meifang Wang
- Department of Pulmonary and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Lisha Zhang
- Department of Obstetrics, Tangshan Caofeidian District Hospital, Tangshan, Hebei, China
| | - Yanhui Chen
- Department of Neuroscience and Endocrinology, Tangshan Caofeidian District Hospital, Tangshan, Hebei, China
| | - Litao Huang
- Department of Clinical Research Management, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Junjun Sun
- Department of Emergency Surgery, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, , China
| | - Dandan Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Yong Chen
- Department of Radio-Chemotherapy, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
3
|
Cui J, Ou Y, Yue K, Wu Y, Duan Y, Liu G, Chen Z, Wei M, Wang X. Comprehensive characterization of the molecular feature of T cells in laryngeal cancer: evidence from integrated single-cell and bulk RNA sequencing data using multiple machine learning approaches. Ann Med 2025; 57:2477287. [PMID: 40179028 PMCID: PMC11980214 DOI: 10.1080/07853890.2025.2477287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/03/2024] [Accepted: 02/10/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND The clinical relevance of T cell-related molecules at single-cell resolution in laryngeal cancer (LC) has not been clarified. MATERIALS AND METHODS Three LC tissues and matching adjoining normal tissues from the hospital were used to perform 10X single-cell RNA sequencing. Hub T cell-related genes (TCRGs) were detected by applying ten machine learning (ML) techniques based TCGA and GEO databases, which were also utilized to create a prediction model (TCRG classifier) and a multicenter validation model. Lastly, we conducted a comprehensive analysis of the TCRG's correlation with immunological properties. RESULTS The analysis of single-cell RNA-seq data revealed that T cells are the primary components of the tumor microenvironment (TME), are significantly involved in cell differentiation pathways, and play a considerable role in intercellular communication. Based on 10 ML approaches, TCRG classifier were identified to develop and validate. The TCRG classifier exhibited excellent prognostic values with a mean C-index of 0.66 in six cohorts, serving as an independent risk factor (p < 0.01). Additionally, the TCRG exhibited a significant relationship with immune score, immune cell infiltration, immune-associated pathways, immune checkpoint inhibitors, human leukocyte antigen, and immunogenicity. Lastly, IPS, TCIC, TIDE, and IMvigor210 cohort analysis illustrated that the immunotherapy response may be accurately predicted using TCRG. CONCLUSION A TCRG classifier is an excellent resource for predicting a patient's prognosis, potentially guiding the preservation of laryngeal function, and identifying patients who may have a positive response to immunotherapy, which might have profound effects on therapeutic practice.
Collapse
Affiliation(s)
- Jie Cui
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, PR China
| | - Yangpeng Ou
- Department of Oncology, Huizhou Third People’s Hospital, Guangzhou Medical University, Huizhou, PR China
| | - Kai Yue
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, PR China
| | - Yansheng Wu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, PR China
| | - Yuansheng Duan
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, PR China
| | - Genglong Liu
- School of Medicine, Southern Medical University, Foshan, PR China
- Editor Office, iMeta, Shenzhen, PR China
| | - Zhen Chen
- Department of Intensive Care Unit, Shunde Hospital, Southern Medical University (the First People’s Hospital of Shunde), Foshan, PR China
| | - Minghui Wei
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, PR China
| | - Xudong Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, PR China
| |
Collapse
|
4
|
Zhang C, Wang Y, Yu Y, Pang Y, Xiao X, Hao L. Overexpression of ST8Sia1 inhibits tumor progression by TGF-β1 signaling in rectal adenocarcinoma and promotes the tumoricidal effects of CD8 + T cells by granzyme B and perforin. Ann Med 2025; 57:2439539. [PMID: 39656552 PMCID: PMC11633436 DOI: 10.1080/07853890.2024.2439539] [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: 02/09/2024] [Revised: 05/23/2024] [Accepted: 10/29/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Rectal adenocarcinoma (READ) involves the dysregulated expression of alpha 2,8-Sialyltransferase1 (ST8Sia1) although its role during READ's progression is unclear. METHODS The mRNA level of ST8Sia1 was analyzed based on The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and Tumor Immune Estimation Resource (TIMER) 2.0. Furthermore, the prognostic and significance of ST8Sia1 in READ was assessed through Kaplan-Meier curve, univariate, multivariate Cox regression, and receiver operating characteristic (ROC) methods. The role of ST8Sia1 in the READ immune microenvironment was explored using ESTIMATE analysis and TIMER databases. Furthermore, the expression of ST8Sia1 in tissues was analyzed using real-time quantitative polymerase chain reaction (RT-qPCR), western blotting (WB), and immunohistochemistry (IHC). Perforin and Granzyme B secretion by CD8+ T cells, as well as tumor cell apoptosis, were detected after co-culturing CD8+ T cells with READ tumor cells and ST8Sia1-overexpression (ST8Sia1-OE) tumor cells. Furthermore, we examined the interaction between ST8Sia1 and TGF-β1 in READ cells. RESULTS ST8Sia1 exhibited excellent diagnostic capability for READ, with positive correlations to immune response and negative correlations to tumor purity. Increased levels of perforin and Granzyme B from CD8+ T cells were observed in vitro, enhancing tumor cell apoptosis. ST8Sia1 interacts with TGF-β1, mediating its inhibitory effects on READ development. CONCLUSIONS ST8Sia1 is a potential diagnostic biomarker and therapeutic target for READ, enhancing CD8+ T cell function and possibly improving patient outcomes through cellular immunotherapy.
Collapse
Affiliation(s)
- Chang Zhang
- Department of Anorectal, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province, China
| | - Yeli Wang
- Department of Anorectal, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province, China
| | - Yao Yu
- Department of General Pediatric Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province, China
| | - Yanchao Pang
- Department of Anorectal, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province, China
| | - Xiao Xiao
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province, China
| | - Leilei Hao
- Department of Anorectal, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province, China
| |
Collapse
|
5
|
Burzinskis E, Janulaityte I, Jievaltas M, Skaudickas D, Burzinskiene G, Dainius E, Naudziunas A, Vitkauskiene A. Inflammatory markers in prostate cancer: potential roles in risk stratification and immune profiling. J Immunotoxicol 2025; 22:2497776. [PMID: 40296239 DOI: 10.1080/1547691x.2025.2497776] [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: 03/10/2025] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
Inflammation plays a critical role in prostate cancer (PCa) pathophysiology, yet the diagnostic value of specific inflammatory markers remains unclear. This study evaluates the association between circulating and tissue inflammatory markers with PCa presence and their potential as biomarkers for risk stratification. This prospective study analyzed serum and prostate biopsy samples from 60 patients with PCa and 22 cancer-free controls. Concentrations of inflammatory markers, including IL-2, IL-4, IL-10, IL-13, IL-33, Oncostatin M, TNFα, PDGF-BB, and TREM-1, were measured using Luminex technology. Statistical analyses included the Mann-Whitney test, logistic regression, and ROC curve analysis to assess differences and diagnostic performance. PCa patients exhibited significantly higher serum levels of IL-2 (p = 0.001), IL-10 (p < 0.001), IL-33 (p < 0.001), Oncostatin M (p = 0.018), and TNFα (p = 0.017) compared to controls. In contrast, biopsy tissue levels of IL-4 (p < 0.001), IL-10 (p < 0.001), IL-13 (p = 0.004), Oncostatin M (p = 0.012), PDGF-BB (p = 0.039), and TREM-1 (p = 0.013) were significantly lower in PCa patients, suggesting an inverse association. IL-10 (inverse) and IL-4 (inverse) in biopsy tissue showed high specificity in ROC analysis (AUC = 0.788 and 0.804, respectively), while IL-2 and IL-33 in serum were positively associated with PCa risk. This study suggests that IL-4, IL-10, and IL-13 in biopsy tissue may serve as biomarkers of a protective effect, while elevated IL-2 and IL-33 in serum are associated with an increased risk of PCa. These findings highlight the potential of inflammatory markers in PCa risk stratification, warranting further investigation in larger cohorts.
Collapse
Affiliation(s)
- Edgaras Burzinskis
- Department of Laboratory Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ieva Janulaityte
- Department of Laboratory Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Mindaugas Jievaltas
- Department of Urology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Darijus Skaudickas
- Department of Urology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Guoda Burzinskiene
- Department of Obstetrics and Gynecology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Edvinas Dainius
- Department of Surgery, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Albinas Naudziunas
- Lithuanian, University of Health Sciences, Internal Medicine Department, Kaunas, Lithuania
| | - Astra Vitkauskiene
- Department of Laboratory Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
6
|
Chouaid C, Grossi F, Ta Thanh Minh C, Raymond R, Bosch-Barrera J. Pooled analysis of oral vinorelbine as single agents in patients with advanced NSCLC. Lung Cancer Manag 2025; 14:2477418. [PMID: 40116568 PMCID: PMC11938966 DOI: 10.1080/17581966.2025.2477418] [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: 06/21/2024] [Accepted: 03/06/2025] [Indexed: 03/23/2025] Open
Abstract
OBJECTIVES This was a pooled analysis of data from weekly vinorelbine (VNR) treatment arms of four individual open-label, phase II studies to assess and refine the efficacy and tolerance of weekly oral VNR in a larger cohort of patients with advanced NSCLC. MATERIALS AND METHODS All patients included in this pooled analysis received oral VNR at the dose of 60 mg/m2 weekly at cycle 1 (3-week cycle), followed by an increase to 80 mg/m2 weekly for subsequent cycles until disease progression or toxicity. Efficacy was based on objective response rate (ORR), progression-free survival (PFS), and disease control rate (DCR). RESULTS A total of 247 patients were included. The ORR and DCR were 8.9% and 57.5% respectively, median PFS and OS were 3.3 and 8.5 months, respectively. Less than half (40.7%) of patients reported ≥1 serious AE (regardless of causality), with 12.3% reporting ≥1 treatment-related serious AE (grade ≥3: 11.1%). The most reported grade ≥3 AEs were neutropenia (17.6%), fatigue (5.8%), and decreased appetite (4.9%). CONCLUSION This pooled analysis showed that weekly oral VRN is a valid option, with an acceptable safety profile, in this population of patients with advanced NSCLC, confirming results from previous individual studies.
Collapse
Affiliation(s)
| | - Francesco Grossi
- Department of Medicine and Technological Innovation, Università degli Studi dell’Insubria, Varese - Medical Oncology Division, ASST Sette Laghi, Varese, Italy
| | | | - Romain Raymond
- Medical & Patient/Consumer Division, Pierre Fabre, Boulogne-Billancourt, France
| | - Joaquim Bosch-Barrera
- Department of Medical Oncology, Catalan Institute of Oncology, Doctor Josep Trueta University Hospital; Precision Oncology Group (OncoGIR-Pro), Institut d’Investigació Biomèdica de Girona (IDIBGI); Department of Medical Sciences, Medical School, University of Girona, Girona, Spain
| |
Collapse
|
7
|
Zhu X, Ma X, Li H, Zhang M, Cheng Y, Wu J, Yu W, Feng W, Zhao L, Li Z, Fu X, Liu J. The efficacy and safety of anlotinib plus PD-1 inhibitor in locally advanced/metastatic esophageal squamous cell carcinoma (ESCC) patients who progressed on prior immune checkpoint inhibitors (ICIs): a retrospective real-world study (NCT 04984096). Ann Med 2025; 57:2443811. [PMID: 39711430 DOI: 10.1080/07853890.2024.2443811] [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: 06/25/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Several studies have shown that combining immune checkpoint inhibitors (ICIs) with antiangiogenic tyrosine kinase inhibitors is effective for solid tumors, including esophageal squamous cell carcinoma (ESCC). However, most of these studies were focused on immunotherapy-naive patients. This retrospective real-world study offers insights into the efficacy and safety of combining anlotinib with ICIs in locally advanced/metastatic ESCC patients who progressed on prior ICI. METHODS We retrospectively analyzed the efficacy and safety of anlotinib plus PD-1 inhibitor in locally advanced/metastatic ESCC patients who had progressed on PD-1 inhibitor. Efficacy was assessed according to the Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1). The primary endpoints were the objective response rate (ORR) and disease control rate (DCR), while secondary endpoints were safety, overall survival (OS) and progression-free survival (PFS). Baseline characteristics and adverse events (AEs) were documented throughout the study. RESULTS Between July 2020 and March 2022, 29 eligible patients were included in the final analysis, with 23 (79.3%) having previously undergone resection for ESCC. Of these 29 patients, 8 (27.6%) received first-line systemic therapy, 20 (69.0%) received second-line therapy, and 1 patient (3%) received third-line therapy. At the data cutoff, the ORR was 31.0%, and the DCR was 86.2%, with 9 patients achieving partial response (PR), 16 patients with stable disease (SD) and 4 patients with disease progression (PD). The median PFS was 5.33 months (95% CI: 4.28-6.38), and the median OS was 10.37 months (95% CI: 6.26-14.46). All patients experienced treatment-related adverse events (TRAEs), with anemia and lymphopenia being the most common. Only 2 patients (6.9%) experiencing grade 3-4 lymphopenia. All AEs were managed with symptomatic treatment and no treatment-related deaths occurred. CONCLUSION The combination of anlotinib and a PD-1 inhibitor demonstrated promising antitumor efficacy and manageable toxicity in patients with locally advanced/metastatic ESCC who progressed on prior ICI. This regimen represents a feasible and well-tolerated treatment option for this patient population. TRIAL REGISTRATION NUMBER NCT04984096.
Collapse
Affiliation(s)
- Xueru Zhu
- Department of Radiation Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiumei Ma
- Department of Radiation Oncology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongxuan Li
- Department of Radiation Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Zhang
- Department of Integrative Medicine, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Cheng
- Department of Radiation Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianguo Wu
- Department of Radiation Oncology, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Wen Yu
- Department of Radiation Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Feng
- Department of Radiation Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Zhao
- Department of Radiation Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhigang Li
- Department of Thoracic Surgery, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolong Fu
- Department of Radiation Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Liu
- Department of Radiation Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Ryu DS, Lee H, Eo SJ, Kim JW, Kim Y, Kang S, Noh JH, Lee S, Park JH, Na K, Kim DH. Photo-responsive self-expanding catheter with photosensitizer-integrated silicone-covered membrane for minimally invasive local therapy in malignant esophageal cancer. Biomaterials 2025; 320:123265. [PMID: 40121828 DOI: 10.1016/j.biomaterials.2025.123265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Photodynamic therapy (PDT) using photosensitizer (PS)-integrated covered self-expandable metallic stents (SEMS) is proposed a new therapeutic approach for the treatment of palliative malignancies; however, the currently hydrophobic PS reduces the photoreactive effect, which leads to aggregation with low water solubility. In here, an aluminum (III)-phthalocyanine chloride tetrasulfonic acid (Al-PcS4)-integrated silicone-covered self-expanding catheter was successfully fabricated to perform localized PDT. The ratio of MeOH and Al-PcS4 concentrations was optimized to achieve PS coating uniformity. The photodynamic activity of the Al-PcS4-integrated silicone membrane was evaluated through laser exposure on membrane-layered tumor cell lines, tumor xenograft-bearing mice. PDT with the Al-PcS4-integrated membrane successfully generated sufficient cytotoxic singlet oxygen, inducing cell death in the esophageal cancer cell lines. PDT-treated tumor xenograft-bearing mice undergo apoptotic cell death and showed significant tumor regression. Localized PDT using an Al-PcS4-integrated silicone-covered self-expanding catheter was technically successful in the rabbit esophagus without severe complications. Based on the endoscopy, esophagography, histology, and immunohistochemistry, our study verified that localized PDT using the Al-PcS4-integrated silicone-covered self-expanding catheter was effective and safe to evenly induce tissue damage. Al-PcS4-integrated silicone-covered self-expanding catheter has substantial potential for the minimally invasive local therapy in malignant esophageal cancer.
Collapse
Affiliation(s)
- Dae Sung Ryu
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea; Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hyeonseung Lee
- Department of Biotechnology, Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Seung Jin Eo
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea; Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Ji Won Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea; Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Yuri Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Seokin Kang
- Department of Internal Medicine, Ilsan Paik Hospital, Inje University College of Medicine, 170, Juhwa-ro, Ilsanseo-gu, Goyang, Gyeonggi-do, 10380, Republic of Korea
| | - Jin Hee Noh
- Department of Internal Medicine, University of Hallym College of Medicine, Hallym University Sacred Heart Hospital, Anyang, Gyeonggi-do, 14068, Republic of Korea
| | - Sanghee Lee
- Department of Radiology Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jung-Hoon Park
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea; Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Kun Na
- Department of Biotechnology, Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea.
| | - Do Hoon Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
9
|
Song H, Kim D, Jang SJ, Hwang HS, Song JS. Clinicopathologic features of histologic transformation in lung adenocarcinoma after treatment with epidermal growth factor receptor-tyrosine kinase inhibitors. Ann Diagn Pathol 2025; 77:152478. [PMID: 40215564 DOI: 10.1016/j.anndiagpath.2025.152478] [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: 03/12/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) may lead to drug resistance, and the underlying mechanism may involve histologic transformations to small cell carcinoma (SCC), squamous cell carcinoma (SqCC), and sarcomatoid carcinoma (SC). Although there are reports regarding these transformations, comprehensive analyses are limited. METHODS A total of 233 patients with primary lung adenocarcinoma treated with EGFR-TKIs were reviewed. Among them, 26 patients (11.1 %) showed histologic transformation. RESULTS Eleven patients (42.3 %) showed SCC and SqCC transformations respectively, and four patients (15.4 %) showed SC transformation. The median time from TKI initiation to transformation was 19.8 months (6.8-51.4) for SCC, 45.3 months (2.4-101.5) for SqCC, and 11.8 months (6.8-15.7) for SC. The median overall survival (OS) was 41.8 months (12.5-78.9), 72.6 months (18.8-112.7), and 23.7 months (17.4-34.4), respectively. The survival from transformation was 12.3 months (2.1-28.3), 16.9 months (0.7-43.2), and 11.4 months (1.6-23.5), respectively. The most common mutations were TP53, PTEN, and RB1 in SCC; TP53 and RB1 in SqCC; and TP53 and KMT2D in SC. SC transformation had the worst OS, followed by SCC and SqCC (p < 0.001). This prognosis difference was also reflected in the time to transformation after EGFR-TKI treatment (p = 0.005). However, survival after transformation was not associated with tumor subtypes (p = 0.536). CONCLUSIONS The analysis of mutation profiles and survival outcomes revealed that the transformation subtype affects prognosis. Additionally, the time taken to undergo transformation is critical for patient outcomes.
Collapse
Affiliation(s)
- Halim Song
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea
| | - Deokhoon Kim
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea
| | - Se Jin Jang
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea
| | - Hee Sang Hwang
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea
| | - Joon Seon Song
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea.
| |
Collapse
|
10
|
Li Y, Liu P, Liu S, Zhu J, Han Y, Jiang Z, Tang D, Meng Z, Li S, Zhang M, Fan Y, Fan F, Zhang P, Liu H. Halofuginone targets Serine/Glycine synthesis to reverse epidermal growth factor receptor Tyrosine Kinase inhibitor resistance in lung adenocarcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156788. [PMID: 40354707 DOI: 10.1016/j.phymed.2025.156788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 04/10/2025] [Accepted: 04/18/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND An emerging issue is that patients are prone to become resistance to epidermal growth factor tyrosine kinase inhibitors (EGFR-TKIs) which are the first- line treatment for EGFR-mutated non-small cell lung cancer (NSCLC) after approximately 10 months of drug administration. Interestingly, metabolic dysregulation occurs simultaneously with acquired EGFR-TKI resistance in certain NSCLC cell lines. PURPOSE We aimed to investigate whether a natural product, halofuginone (HF), could overcome NSCLC resistance to EGFR-TKIs by influencing metabolism. RESULTS In our study, the combination of HF and EGFR-TKI exhibited synergistic cytotoxicity compared to EGFR-TKI monotherapy. The underlying mechanism is that HF promotes the degradation of SP1 protein and decreases the expression of phosphatidylserine transcarbamoylase 1 (PSAT1), which leads to defects in the de novo synthesis of Serine/Glycine and cell death. CONCLUSIONS HF is a promising natural product for overcoming NSCLC resistance to third-generation epidermal growth factor receptors-TKIs.
Collapse
Affiliation(s)
- Yongping Li
- School of Pharmacy, Bengbu Medical University, Bengbu, 233000, China
| | - Peipei Liu
- School of Pharmacy, Bengbu Medical University, Bengbu, 233000, China
| | - Shiyao Liu
- School of Pharmacy, Bengbu Medical University, Bengbu, 233000, China
| | - Juan Zhu
- School of Pharmacy, Bengbu Medical University, Bengbu, 233000, China
| | - Yuehua Han
- School of Pharmacy, Bengbu Medical University, Bengbu, 233000, China
| | - Zuojie Jiang
- School of Pharmacy, Bengbu Medical University, Bengbu, 233000, China
| | - Dawei Tang
- School of Pharmacy, Bengbu Medical University, Bengbu, 233000, China
| | - Zixuan Meng
- School of Pharmacy, Bengbu Medical University, Bengbu, 233000, China
| | - Shanshan Li
- School of Pharmacy, Bengbu Medical University, Bengbu, 233000, China; Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, 233000, China
| | - Mengxiao Zhang
- School of Pharmacy, Bengbu Medical University, Bengbu, 233000, China; Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, 233000, China
| | - Yunlei Fan
- School of Pharmacy, Bengbu Medical University, Bengbu, 233000, China
| | - Fangtian Fan
- School of Pharmacy, Bengbu Medical University, Bengbu, 233000, China; Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, 233000, China
| | - Pei Zhang
- School of Pharmacy, Bengbu Medical University, Bengbu, 233000, China; Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, 233000, China
| | - Hao Liu
- School of Pharmacy, Bengbu Medical University, Bengbu, 233000, China; Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, 233000, China.
| |
Collapse
|
11
|
Linssen JDG, Schafrat PJM, de Back TR, van Erning FN, van Leerdam ME, Dekker E, Vermeulen L, de Hingh IHJT, Sommeijer DW. Predisposing conditions in patients with small intestinal adenocarcinomas in the Netherlands: A 20-year nationwide cohort study. Int J Cancer 2025; 157:218-231. [PMID: 39907526 PMCID: PMC12079630 DOI: 10.1002/ijc.35354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/06/2025]
Abstract
Small intestinal adenocarcinomas (SIAs) are associated with predisposing conditions, including inflammatory bowel disease (IBD) and celiac disease, but also genetic syndromes such as Lynch syndrome (LS) and familial adenomatous polyposis (FAP). This nationwide cohort study investigated the incidence of genetic and non-genetic predisposing conditions in SIA and their influence on tumor characteristics and clinical features. Data were obtained from the Netherlands Cancer Registry. The incidence, characteristics, and clinical features per predisposing condition were analyzed in 2697 SIA patients diagnosed from 1999 through 2019. Of all SIA patients, 5.6% were known to have a genetic predisposing syndrome, of whom 4.0% had LS and 1.6% had a polyposis syndrome. In addition, 6.8% of SIA patients had a non-genetic predisposing condition: 3.9% IBD and 2.9% celiac disease. SIAs of patients with such predisposing syndromes or conditions were diagnosed at a younger age and earlier stage and affected the duodenum less often as compared to sporadic SIA patients. Both genetic and non-genetic predisposing conditions were associated with significantly better overall survival (OS) compared to sporadic SIA: sporadic SIA (median OS: 13.0 months, 95% CI: 11.8-14.2), LS (213.1 months, 99.3-NA), polyposis syndromes (61.3 months, 19.7-NA), IBD (29.5 months, 20.3-69.8), and celiac disease (50.4 months, 24.6-124.7). This nationwide cohort study shows significant differences between SIA with and without predisposing conditions and highlights the need for research on underlying molecular mechanisms to improve outcomes of SIA patients.
Collapse
Affiliation(s)
- Jasmijn D. G. Linssen
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and RadiobiologyCenter for Experimental and Molecular MedicineAmsterdamThe Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory for Experimental Oncology and RadiobiologyCenter for Experimental and Molecular MedicineAmsterdamThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
- Department of Gastroenterology and HepatologyAmsterdam UMC, Location University of AmsterdamAmsterdamThe Netherlands
| | - Pascale J. M. Schafrat
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and RadiobiologyCenter for Experimental and Molecular MedicineAmsterdamThe Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory for Experimental Oncology and RadiobiologyCenter for Experimental and Molecular MedicineAmsterdamThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
- Department of Medical OncologyAmsterdam UMC, Location University of AmsterdamAmsterdamThe Netherlands
| | - Tim R. de Back
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and RadiobiologyCenter for Experimental and Molecular MedicineAmsterdamThe Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory for Experimental Oncology and RadiobiologyCenter for Experimental and Molecular MedicineAmsterdamThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Felice N. van Erning
- Department of SurgeryCatharina HospitalEindhovenThe Netherlands
- Department of Research and DevelopmentNetherlands Comprehensive Cancer Organization (IKNL)UtrechtThe Netherlands
| | - Monique E. van Leerdam
- Department of Gastroenterology and HepatologyLeiden University Medical CenterLeidenThe Netherlands
- Department of Gastrointestinal OncologyNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Evelien Dekker
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and RadiobiologyCenter for Experimental and Molecular MedicineAmsterdamThe Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory for Experimental Oncology and RadiobiologyCenter for Experimental and Molecular MedicineAmsterdamThe Netherlands
- Department of Gastroenterology and HepatologyAmsterdam UMC, Location University of AmsterdamAmsterdamThe Netherlands
| | - Louis Vermeulen
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and RadiobiologyCenter for Experimental and Molecular MedicineAmsterdamThe Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory for Experimental Oncology and RadiobiologyCenter for Experimental and Molecular MedicineAmsterdamThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Ignace H. J. T. de Hingh
- Department of SurgeryCatharina HospitalEindhovenThe Netherlands
- Department of Research and DevelopmentNetherlands Comprehensive Cancer Organization (IKNL)UtrechtThe Netherlands
- Department of Epidemiology, GROW‐School for Oncology and Developmental BiologyMaastricht UniversityMaastrichtThe Netherlands
| | - Dirkje W. Sommeijer
- Cancer Center Amsterdam, Laboratory for Experimental Oncology and RadiobiologyCenter for Experimental and Molecular MedicineAmsterdamThe Netherlands
- Department of Internal MedicineFlevohospitalAlmereThe Netherlands
| |
Collapse
|
12
|
Reddyrajula R, Kathirvel PV, Shankaraiah N. Recent developments of benzimidazole based analogs as potential tubulin polymerization inhibitors: A critical review. Bioorg Med Chem Lett 2025; 122:130167. [PMID: 40074012 DOI: 10.1016/j.bmcl.2025.130167] [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: 01/27/2025] [Revised: 02/18/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Microtubules, as dynamic regulators in many cellular processes, remain pivotal targets in cancer chemotherapy. Among the structural motifs explored, the benzimidazole scaffold has emerged as a privileged heterocyclic ring system in the development of potent therapeutic agents, owing to its versatility and pharmacological relevance. This review critically examines the synthesis, anticancer activity, structure-activity relationships (SAR), and tubulin polymerization inhibitory properties of diverse benzimidazole derivatives. In addition, various synthetic strategies and innovative approaches for generating benzimidazole based analogs with enhanced cytotoxic profiles are highlighted. Recent findings underscore the potential of benzimidazole derivatives as promising tubulin polymerization inhibitors, contributing significantly to the discovery of next-generation anticancer agents.
Collapse
Affiliation(s)
- Rajkumar Reddyrajula
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad-500037, India
| | - Priya Varshini Kathirvel
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad-500037, India
| | - Nagula Shankaraiah
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad-500037, India.
| |
Collapse
|
13
|
Xu X, Wang L, Cao Y, Wang J, Yao J, Huo L, Song Y, Ding W, Gu J, Li Q. Long-term follow-up outcomes of intraoperative radiotherapy for breast-conserving treatment in early breast cancer: A retrospective cohort study. Oncol Lett 2025; 30:346. [PMID: 40438870 PMCID: PMC12117530 DOI: 10.3892/ol.2025.15092] [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: 11/01/2024] [Accepted: 04/17/2025] [Indexed: 06/01/2025] Open
Abstract
Intraoperative radiotherapy (IORT) has been used as a novel therapeutic alternative for breast-conserving surgery (BCS) in patients with breast cancer. However, the long-term outcomes and safety of IORT in patients with breast cancer remain incompletely understood. Therefore, the present study aimed to explore the long-term outcomes of IORT following BCS, focusing on the oncological outcomes and cosmetic consequences compared with postoperative RT (PORT) in patients with breast cancer. Patients with early-stage breast cancer who underwent BCS followed by IORT or PORT between January 2016 and October 2020 at the Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University (Nanjing, China), were retrospectively reviewed. After screening, a total of 59 patients were included in the present study and divided into two groups according RT records as follows: The IORT group (n=21) and the PORT group (n=38). The clinical data of all patients, including surgical and RT complications, cosmetic grading and scoring, and other events, were collected and retrospectively analyzed. No significant difference was observed in terms of the mean follow-up time in the IORT (5.89±1.57 years) and PORT (6.09±1.60 years) groups (P>0.05). Compared with PORT, IORT showed non-inferior therapeutic efficacy, with no significant differences in postoperative complications (surgical site infection and chronic pain; P>0.05). Notably, the IORT group achieved superior cosmetic outcomes, with 95.2% (20/21) of patients achieving a rating of excellent/good vs. 44.7% (17/38 patients) in the PORT group (P<0.001), alongside higher median cosmetic scores at all postoperative intervals (P<0.01). IORT also reduced healthcare utilization, shortening hospitalization by 23.8 days (12.1±5.1 vs. 35.9±3.5 days; P<0.001) and lowering costs significantly (33,117.98±6,281.17 vs. 77,789.55±7,000.90 CNY; P<0.001). These findings suggest that IORT offers comparable safety, improved cosmesis and greater cost-effectiveness than PORT, supporting its integration into clinical practice for eligible patients.
Collapse
Affiliation(s)
- Xiaofan Xu
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu 210002, P.R. China
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Lu Wang
- Research Institute of General Surgery, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210002, P.R. China
| | - Yuanyuan Cao
- Medical Service Training Center, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Jingyi Wang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Jie Yao
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu 210002, P.R. China
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Liqun Huo
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu 210002, P.R. China
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Yuqing Song
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Wei Ding
- Department of Radiation Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Jun Gu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Qiurong Li
- Research Institute of General Surgery, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu 210002, P.R. China
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
14
|
Wang W, Liu X, Xu S, Dai E, Li Y, Liu Y, Shan L, Li Y. CD38 contributes to tumor progression and tumor microenvironment reshaping in epithelial ovarian cancer. Transl Oncol 2025; 57:102414. [PMID: 40381484 PMCID: PMC12143800 DOI: 10.1016/j.tranon.2025.102414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 04/08/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Ovarian cancer, ranking fifth in cancer mortality, presents a significant therapeutic challenge. The immunomodulatory functions of CD38in epithelial ovarian cancer (EOC) and its influence on the tumor microenvironment (TME) remain poorly understood. METHODS Public datasets, RT-qPCR and immunohistochemistry (IHC) were used to analyze CD38 expression and clinicopathological features in EOC. Gene manipulation techniques were employed to elucidate its functions, while integrated IHC and bioinformatics were conducted to assess its involvement in immune/stromal infiltration. Immune-related functions of CD38 were explored using GO, KEGG analysis and TIP database. TIDE algorithm was employed to predict the correlation between CD38 and immune checkpoint blocking responsiveness. CD38 inhibitor efficacy was evaluated in an EOC mouse model, with flow cytometry monitoring cellular changes. The involvement of CD38 in the PI3K-AKT and IL-6 signaling pathways was evaluated using RT-qPCR, western blot, and publicly datasets. RESULTS CD38 is significantly upregulated in EOC, influencing the cell proliferation and metastasis. It regulates the PI3K-AKT and IL-6 signaling pathways, thereby increasing tumor malignancy. CD38 is also upregulated in immune and stromal cells, affecting TME remodeling by facilitating immune cell and CAF infiltration, impeding T cell recognition of tumor cells, and enhancing CAF-tumor cell communication. Additionally, CD38 correlates with multiple immune checkpoint molecules. Notably, CD38 inhibitor therapy inhibited effectively EOC progression and modulates immune responses. CONCLUSION Elevated CD38 expression is associated with EOC progression, TME remodeling, and immune response modulation. Thus, CD38 could be a promising target for ovarian cancer immunotherapy.
Collapse
Affiliation(s)
- Wei Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| | - Xiangnan Liu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| | - Shengjie Xu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| | - Enci Dai
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| | - Yingying Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| | - Yinping Liu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| | - Liyun Shan
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| | - Yanli Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
15
|
Qin R, Tang Y, Yuan Y, Meng F, Zheng K, Yang X, Zhao J, Yang C. Studies on the functional role of UFMylation in cells (Review). Mol Med Rep 2025; 32:191. [PMID: 40341950 PMCID: PMC12076054 DOI: 10.3892/mmr.2025.13556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/16/2025] [Indexed: 05/11/2025] Open
Abstract
Protein post‑translational modifications (PTMs) play crucial roles in various life activities and aberrant protein modifications are closely associated with numerous major human diseases. Ubiquitination, the first identified protein modification system, involves the covalent attachment of ubiquitin molecules to lysine residues of target proteins. UFMylation, a recently discovered ubiquitin‑like modification, shares similarities with ubiquitination. The precursor form of ubiquitin fold modifier 1 (UFM1) undergoes synthesis and cleavage by UFM1‑specific protease 1 or UFM1‑specific protease 2 to generate activated UFM1‑G83. Subsequently, UFM1‑G83 is activated by a specific E1‑like activase, UFM1‑activating enzyme 5. UFM1‑conjugating enzyme 1 and an E3‑like ligase, UFM1‑specific ligase 1, recognize the target protein and facilitate UFMylation, leading to the degradation of the target protein. Current knowledge regarding UFMylation remains limited. Previous studies have demonstrated that defects in the UFMylation pathway can result in embryonic lethality in mice and various human diseases, highlighting the critical biological functions of UFMylation. However, the precise mechanisms underlying UFMylation remain elusive. This present review aimed to summarize recent research advances in UFMylation, with the aim of providing novel insights and perspectives for future investigations into this essential protein modification system.
Collapse
Affiliation(s)
- Rong Qin
- Yunan Key Laboratory of Breast Cancer Precision Medicine, School of Biomedical Engineering, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yu Tang
- Yunan Key Laboratory of Breast Cancer Precision Medicine, The Third Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yuhang Yuan
- Yunan Key Laboratory of Breast Cancer Precision Medicine, School of Biomedical Engineering, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Fangyu Meng
- Yunan Key Laboratory of Breast Cancer Precision Medicine, School of Biomedical Engineering, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Kepu Zheng
- Department of Hepato-Biliary-Pancreatic Surgery, The Affiliated Calmette Hospital of Kunming Medical University, The First People's Hospital of Kunming, Kunming, Yunnan 650000, P.R. China
| | - Xingyu Yang
- Yunan Key Laboratory of Breast Cancer Precision Medicine, The Third Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Jiumei Zhao
- Department of Laboratory, Chongqing Nanchuan District People's Hospital, Chongqing Medical University, Chongqing 408400, P.R. China
| | - Chuanhua Yang
- Department of General Surgery, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
16
|
Hashemi M, Gholamrezaie H, Ziyaei F, Asadi S, Naeini ZY, Salimian N, Enayat G, Sharifi N, Aliahmadi M, Rezaie YS, Khoushab S, Rahimzadeh P, Miri H, Abedi M, Farahani N, Taheriazam A, Nabavi N, Entezari M. Role of lncRNA PVT1 in the progression of urological cancers: Novel insights into signaling pathways and clinical opportunities. Cell Signal 2025; 131:111736. [PMID: 40081549 DOI: 10.1016/j.cellsig.2025.111736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/02/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Urologic malignancies, encompassing cancers of the kidney, bladder, and prostate, represent approximately 25 % of all cancer cases. Recent advances have enhanced our understanding of PVT1's crucial functions. Long noncoding RNAs influence both the onset and development of cancer, as well as epigenetic alterations. Recent findings have focused on PVT1's mechanism of action across several malignancies, particularly urologic cancers. Understanding the various functions of PVT1 linked to cancer is necessary for the development of cancer detection and treatment when PVT1 is dysregulated. Furthermore, recent advancements in genomic and epigenetic research have elucidated the complex regulatory networks that control PVT1 expression. Comprehending the intricate role of PVT1 Understanding the complex function of PVT1 in urologic cancers has substantial clinical implications. Here, we summarize some of the most recent findings about the carcinogenic effects of PVT1 signaling pathways and the possible treatment strategies for urological malignancies that target these pathways.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamidreza Gholamrezaie
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Faezeh Ziyaei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Yousefian Naeini
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Cellular and Molecular Biology,Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Niloufar Salimian
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Golnaz Enayat
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nafiseh Sharifi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Melika Aliahmadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yasamin Soofi Rezaie
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saloomeh Khoushab
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hossein Miri
- Faculty of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Abedi
- Department of Pathology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran,Iran.
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
17
|
Netaji A, Kubihal V, Sharma R, Seth A, Kaushal S, Das CJ. A Prospective Comparison of MRI-Guided Targeted Biopsy with 12-Core Transrectal Ultrasound-Guided Systematic Biopsy in the Diagnosis of Clinically Significant Prostate Cancer: An Indian Experience. Indian J Radiol Imaging 2025; 35:387-394. [PMID: 40529983 PMCID: PMC12169941 DOI: 10.1055/s-0044-1796642] [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] [Indexed: 06/20/2025] Open
Abstract
Objective The aim of this study was to compare the sensitivity and prostate cancer detection rate of magnetic resonance (MR) in-bore biopsy with transrectal ultrasound (TRUS) guided systematic biopsy. We also compared the cancer detection rate of the combined MR in-bore and TRUS-guided systematic biopsy with the TRUS-guided biopsy only approach. Materials and Methods In this prospective study, 61 consecutive patients with prostate-specific antigen (PSA) ≥ 3 ng/mL and Prostate Imaging Reporting and Data System (PI-RADS) score ≥4 were recruited between July 2017 and January 2020. One patient with prior prostate surgery was excluded. Among the remaining 60 patients, 30 underwent MR in-bore biopsy followed by systematic biopsy (study arm A) and 30 underwent systematic biopsy only (study arm B). Results The mean PSA range of study population ( n = 60 patients) was 4.2 to 72.7 ng/mL. Twenty-seven patients had a PI-RADS score of 4, and 33 patients had a PI-RADS score of 5. Among 60 patients, 30 had prostate carcinoma on biopsy, of which 18 were clinically significant prostate cancers (csPCa). In study arm A, TRUS-guided systematic biopsy had a lower sensitivity (0.9) for detection of csPCa compared with MR in-bore biopsy (1.0) with overdetection of insignificant cancers (sensitivity: 0.89 vs. 0.56). TRUS-guided biopsy yielded 112 positive cores out of 360, whereas MR in-bore biopsy yielded 15 positive cores out of 30 (31.1 vs. 50%; p = 0.03). On comparison of study arms A and B, the diagnostic yield for detection of both prostate cancer and csPCa were high in study arm A (60 vs. 40%, and 33.3 vs. 26.7%, respectively) Conclusion MRI in-bore targeted biopsy had a greater sensitivity to detect csPCa with fewer number of biopsy cores and lower sensitivity to detect insignificant cancers compared with systematic biopsy. Systematic biopsies were associated with overdetection of clinically insignificant cancers.
Collapse
Affiliation(s)
- Arjunlokesh Netaji
- Department of Radiology, All India Institute of Medical Sciences, New Delhi, India
| | - Vijay Kubihal
- Department of Radiology, All India Institute of Medical Sciences, New Delhi, India
| | - Raju Sharma
- Department of Radiology, All India Institute of Medical Sciences, New Delhi, India
| | - Amlesh Seth
- Department of Urology, All India Institute of Medical Sciences, New Delhi, India
| | - Seema Kaushal
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Chandan J. Das
- Department of Radiology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
18
|
Cannella R, Dioguardi Burgio M, Maino C, Matteini F, Ippolito D, Boraschi P, Zamboni GA, Vernuccio F. Conditions at risk of pancreatic cancer: The radiology perspective. Eur J Radiol 2025; 188:112119. [PMID: 40273500 DOI: 10.1016/j.ejrad.2025.112119] [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: 11/10/2024] [Revised: 03/28/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
Pancreatic cancer remains one for the most aggressive cancer worldwide, with pancreatic ductal adenocarcinoma being the most common malignant pancreatic lesion, associated with poor prognosis. While surgical resection is the only curative treatment, only a minority of patients is eligible for surgery due to its diagnosis at advanced stages. Therefore, strategies for early detection of pancreatic cancer are needed. This article aims to provide a state-of-the-art review of the most common conditions associated to an increased risk of pancreatic cancer. Conditions linked to risk of pancreatic cancer development include certain pancreato-biliary anatomical variants, intraductal papillary mucinous neoplasms, mucinous cystic neoplasm, and familial pancreatic cancer with specific genetic mutations. Early imaging signs of pancreatic cancer can also be incidentally encountered on CT or MRI performed for other indications and they should be promptly recognized by the radiologists in order to avoid delays in the diagnosis. The features include focal pancreatic atrophy, contour deformity, dilation of the main pancreatic duct (MPD), changes in the caliber of the MPD, abrupt interruption of the MPD, and biliary tree dilation. MRI with the adoption of abbreviated protocols has been increasingly evaluated for the follow-up of cystic lesions. Although screening of the general population is not recommended due to the low incidence and high costs, surveillance with MRI can be considered in selected high-risk individuals.
Collapse
Affiliation(s)
- Roberto Cannella
- Section of Radiology - Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo 90127, Italy.
| | - Marco Dioguardi Burgio
- Université Paris Cité, INSERM, Centre de recherche sur l'inflammation, F-75018 Paris, France; Radiology Department, AP-HP, Hôpital Beaujon, 92110 Clichy, France
| | - Cesare Maino
- Department of Diagnostic Radiology, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi 33, 20900 Monza, MB, Italy
| | - Francesco Matteini
- Section of Radiology - Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo 90127, Italy
| | - Davide Ippolito
- Department of Diagnostic Radiology, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi 33, 20900 Monza, MB, Italy
| | - Piero Boraschi
- 2nd Unit of Radiology, Department of Radiological Nuclear and Laboratory Medicine - Pisa University Hospital, Via Paradisa 2, 56124 Pisa, Italy
| | - Giulia A Zamboni
- Department of Diagnostics and Public Health, Institute of Radiology, University of Verona, Policlinico GB Rossi, P.Le LA Scuro 10, 37134 Verona, Italy
| | - Federica Vernuccio
- Section of Radiology - Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo 90127, Italy
| |
Collapse
|
19
|
Su Y, Leng M, Yang Q, Jiang W, Xiang G, Long L, Zhou X. Targeting circulating tumor cell‒neutrophil interactions: nanoengineered strategies for inhibiting cancer metastasis. J Nanobiotechnology 2025; 23:449. [PMID: 40528159 DOI: 10.1186/s12951-025-03522-8] [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: 03/23/2025] [Accepted: 05/30/2025] [Indexed: 06/20/2025] Open
Abstract
Metastasis remains the leading cause of cancer-related mortality, with a persistently poor prognosis for metastatic cancer patients despite extensive therapeutic efforts. Circulating tumor cells (CTCs), which detach from primary tumors and enter the bloodstream, can establish distant metastatic sites. These CTCs often form heterotypic clusters with white blood cells, especially neutrophils, through various interaction mechanisms, including intercellular adhesion, cytokine secretion, protease release, and the formation of neutrophil extracellular traps (NETs). These interactions enhance CTCs survival, proliferation, invasion, and transendothelial migration while simultaneously remodeling premetastatic niches and the tumor microenvironment. Consequently, pharmacologically disrupting CTC‒neutrophil crosstalk represents a promising strategy to curb metastatic spread and improve clinical outcomes. Recent breakthroughs in nanotechnology-based drug delivery systems have shown considerable potential in antimetastatic therapies, offering significant advantages over conventional treatments, which are often associated with severe side effects and limited efficacy. This review systematically explores nanoengineered strategies targeting CTC‒neutrophil interactions, addresses the current limitations and outlines future directions for developing clinically translatable nanotherapeutics.
Collapse
Affiliation(s)
- Yong Su
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People's Republic of China
| | - Mingjing Leng
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People's Republic of China
| | - Qingqing Yang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People's Republic of China
| | - Wenbi Jiang
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Rehabilitation School, Kunming Medical University, Kunming, 650500, People's Republic of China
| | - Gang Xiang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, People's Republic of China
| | - Ling Long
- School of Pharmaceutical Sciences and Yunnan Provincial Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, People's Republic of China.
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400054, People's Republic of China.
| | - Xing Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Rehabilitation School, Kunming Medical University, Kunming, 650500, People's Republic of China.
| |
Collapse
|
20
|
Meng C, Zang A, Du F. Prognostic value of LncRNA SH3BP5-AS1 in non-small cell lung cancer and its regulatory effect on tumor progression. Discov Oncol 2025; 16:1126. [PMID: 40523970 DOI: 10.1007/s12672-025-02906-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 06/04/2025] [Indexed: 06/19/2025] Open
Abstract
OBJECTIVE This study aimed to investigate the prognostic significance of SH3BP5-AS1 in non-small cell lung cancer (NSCLC) patients, and also to uncover its function in the progression of NSCLC. METHODS 100 NSCLC patients were recruited in this study. SH3BP5-AS1 was measured by RT-qPCR. The prognostic significance of SH3BP5-AS1 in NSCLC progression was appraised utilizing the Kaplan-Meier and Cox regression test. CCK-8 was used to detect the proliferative viability of NSCLC cells. The migratory and invasive capabilities of NSCLC cells were evaluated by Transwell assay. The interplay between SH3BP5-AS1 and miR-424-5p was verified by luciferase reporter assay and ENCORI database. The potential target genes of miR-424-5p were inspected by means of bioinformatics analysis. RESULTS A reduced SH3BP5-AS1 was manifested in tissues of NSCLC patients and tumor cells. Compared with patients showing SH3BP5-AS1 low-expression, those with SH3BP5-AS1 high-expression possessed a more favorable progression-free survival time. SH3BP5-AS1 might function as an autonomous prognostic biomarker for NSCLC. Upregulation of SH3BP5-AS1 suppressed the proliferation, migration and invasion of NSCLC cells. MiR-424-5p was a downstream target miRNA of SH3BP5-AS1, and luciferase reporter assays verified that SH3BP5-AS1 and miR-424-5p interact. In NSCLC patient tissues, miR-424-5p was upregulated and it exhibited an inverse correlation with SH3BP5-AS1. Elevation of miR-424-5p neutralized the inhibitory effect of SH3BP5-AS1 overexpression on the proliferation, migration, and invasion abilities of NSCLC cells. CONCLUSION SH3BP5-AS1 is involved in NSCLC development by targeting its downstream target miRNA miR-424-5p, and it is a latent prognostic indicator for NSCLC.
Collapse
Affiliation(s)
- Cong Meng
- Department of Radiotherapy for Tumors, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Aihua Zang
- Department of Ultrasound, Qingdao Municipal Hospital, Qingdao, 266011, Shandong, China
| | - Fengcai Du
- Department of Oncology, Yantai Yuhuangding Hospital affiliated to Qingdao University (Laishan branch), No.59, Shuanghe West Road, Laishan District, Yantai, 264000, Shandong, China.
| |
Collapse
|
21
|
Lu J, Cai D, Qian L, Wang Y, Ai S, Song P, Sun F, Sun Y, Liang M, Jiang H, Wang X, Wang M, Lu X, Guan W, Shen X. Targeting SIX2 as a novel sensitization strategy of sorafenib treatment on advanced hepatocellular carcinoma through modulating METTL9-SLC7A11 axis. NPJ Precis Oncol 2025; 9:186. [PMID: 40523929 DOI: 10.1038/s41698-025-01004-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 06/09/2025] [Indexed: 06/19/2025] Open
Abstract
Sorafenib is the main treatment for advanced hepatocellular carcinoma (HCC), but drug resistance limits its effectiveness. Evidence increasingly indicates that, in addition to targeting tyrosine kinases, sorafenib also induces ferroptosis. However, current studies have not fully clarified the relationship between ferroptosis and sorafenib treatment sensitivity. Our bioinformatics analysis identified that SIX Homeobox 2 (SIX2), known for maintaining cellular stemness via the Wnt signaling pathway, was significantly upregulated in sorafenib-resistant tissues. Overexpression and knockdown experiments revealed that altering SIX2 expression affected HCC cell sensitivity to sorafenib and involved the ferroptosis pathway, suggesting a regulatory role for SIX2 in ferroptosis. RNA sequencing and CUT&Tag analysis showed that SIX2 directly regulated methyltransferase 9 (METTL9) expression. Co-immunoprecipitation (Co-IP) assays confirmed that METTL9 bound to SLC7A11, enhancing its stability and reducing degradation, thus regulating ferroptosis. Importantly, the role of SIX2 in ferroptosis operated independently of the classical glutathione peroxidase 4 (GPX4) pathway. In vitro studies further supported these findings, demonstrating that SIX2 knockdown increased sorafenib-induced ferroptosis in HCC, while METTL9 overexpression largely counteracted the effects of SIX2 knockdown. In mouse models, overexpression of SIX2 increased tumor resistance to sorafenib. Our findings suggest that modulating the ferroptosis pathway through SIX2 could enhance sorafenib sensitivity. This study provides the first evidence that SIX2 influences ferroptosis via the METTL9-SLC7A11 axis, thereby sensitizing HCC cells to sorafenib. Reducing SIX2 expression could thus represent a promising strategy to improve the efficacy of sorafenib in advanced HCC.
Collapse
Affiliation(s)
- Junren Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of General Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Daming Cai
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Long Qian
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yurong Wang
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shichao Ai
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Peng Song
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Feng Sun
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yiwen Sun
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Mengjie Liang
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hang Jiang
- Department of General Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Xingzhou Wang
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Meng Wang
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Xiaofeng Lu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Wenxian Guan
- State Key Laboratory of Pharmaceutical Biotechnology, Department of General Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of General Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Xuzhou Medical University, Xuzhou, China.
- Department of General Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Xiaofei Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of General Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of General Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
22
|
Yang F, Yang J, Zhu C, Ding T, Zhang X, Zhang H. Olfactory receptor OR51B5 suppressed esophageal cancer progression through activates Calcium / N-Ras signaling. Cell Death Dis 2025; 16:450. [PMID: 40523880 DOI: 10.1038/s41419-025-07769-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 05/14/2025] [Accepted: 06/04/2025] [Indexed: 06/19/2025]
Abstract
The transcriptional regulation of olfactory receptors (ORs) plays a critical role in various biological processes, and has recently been considered a potential therapeutic target for cancer treatment. Esophageal cancer (EC) is a highly invasive neoplasm with dismal prognosis, but the specific roles of ORs in EC remain largely unexplored. Here, we developed a comprehensive workflow to identify potential functional olfactory receptor family 51 subfamily B member 5 (OR51B5) and demonstrated that OR51B5 locus acted as a key spatial element contributing to the progression of esophageal cancer. Moreover, we showed that the CTCF-EZH2 enhanced the trimethylation of lysine 27 of histone H3 (H3K27me3) and increased repressive and closed chromatin state at the OR51B5 promoter region. Subsequently we demonstrated that closed chromatin impaired the entry of RNA polymerase II and inhibited the transcription of OR51B5, thereby causing N-Ras activation and promoting tumor cell proliferation and metastasis. Our study provides an alternative workflow for discovering critical regulatory sites for control tumorigenesis, and reveals a novel OR51B5 triggering mechanism underlying esophageal cancer progression.
Collapse
Affiliation(s)
- Fan Yang
- State Key Laboratory of Cardiology and Medical Innovation Center, Province Key Laboratory of Organ Development and Epigenetics, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, Jinggangshan Enclave Laboratory, School of Life Science and Technology, Tongji University, Shanghai, P. R. China
- Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, P.R. China
- School of Life Science, Jinggangshan University, Ji'an, P.R. China
| | - Jiaqi Yang
- State Key Laboratory of Cardiology and Medical Innovation Center, Province Key Laboratory of Organ Development and Epigenetics, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, Jinggangshan Enclave Laboratory, School of Life Science and Technology, Tongji University, Shanghai, P. R. China
- Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, P.R. China
- School of Life Science, Jinggangshan University, Ji'an, P.R. China
| | - Chengbo Zhu
- State Key Laboratory of Cardiology and Medical Innovation Center, Province Key Laboratory of Organ Development and Epigenetics, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, Jinggangshan Enclave Laboratory, School of Life Science and Technology, Tongji University, Shanghai, P. R. China
- Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, P.R. China
- School of Life Science, Jinggangshan University, Ji'an, P.R. China
| | - Tianyi Ding
- State Key Laboratory of Cardiology and Medical Innovation Center, Province Key Laboratory of Organ Development and Epigenetics, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, Jinggangshan Enclave Laboratory, School of Life Science and Technology, Tongji University, Shanghai, P. R. China
- Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, P.R. China
- School of Life Science, Jinggangshan University, Ji'an, P.R. China
| | - Xiaoyu Zhang
- State Key Laboratory of Cardiology and Medical Innovation Center, Province Key Laboratory of Organ Development and Epigenetics, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, Jinggangshan Enclave Laboratory, School of Life Science and Technology, Tongji University, Shanghai, P. R. China
- Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, P.R. China
- School of Life Science, Jinggangshan University, Ji'an, P.R. China
| | - He Zhang
- State Key Laboratory of Cardiology and Medical Innovation Center, Province Key Laboratory of Organ Development and Epigenetics, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, Jinggangshan Enclave Laboratory, School of Life Science and Technology, Tongji University, Shanghai, P. R. China.
- Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, P.R. China.
- School of Life Science, Jinggangshan University, Ji'an, P.R. China.
| |
Collapse
|
23
|
Zheng M. Elevated CD14 in B cells associates with reduced ovarian cancer risk via CD80 + dendritic cell interaction: a multi-omics study. Discov Oncol 2025; 16:1113. [PMID: 40517355 PMCID: PMC12167730 DOI: 10.1007/s12672-025-02956-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 06/10/2025] [Indexed: 06/18/2025] Open
Abstract
INTRODUCTION Ovarian cancer (OC) is a highly aggressive malignancy characterized by a complex immune microenvironment. B cells, essential components of immunological regulation, have been implicated in the progression of ovarian cancer. However, the precise mechanisms by which B cells and immune molecules influence ovarian cancer risk remain poorly understood. METHODS This study employed single-cell RNA sequencing (scRNA-seq) to analyze peripheral blood mononuclear cells (PBMCs) from ovarian cancer patients and healthy donors. Differential gene expression analysis identified CD14 as a critical gene in B cells. Mendelian randomization (MR) analysis, using exposure data from eQTL and pQTL databases, was performed to evaluate the association between CD14 and ovarian cancer risk. Mediation analysis was conducted to assess the role of CD80 on myeloid dendritic cells in mediating the relationship between CD14 and ovarian cancer. RESULTS The analysis demonstrated that CD14 expression was significantly downregulated in B cells from ovarian cancer patients compared to healthy donors. MR analysis revealed a significant association between elevated CD14 expression and reduced ovarian cancer risk. Mediation analysis indicated that CD80 mediated 26.2% of this effect. CONCLUSION These findings highlight CD14 as a key regulator of ovarian cancer risk, with CD80 serving as a mediator of the immune response in this context. This study provides insights into potential immune modulation strategies for ovarian cancer therapy.
Collapse
Affiliation(s)
- Mengyu Zheng
- Department of Obstetrics and gynecology, Affiliated People's Hospital of NingBo University, 251 Baizhang east Road, Ningbo, 315040, Zhejiang, People's Republic of China.
| |
Collapse
|
24
|
Xie J, Shu X, Xie Z, Tang J, Wang G. Pharmacological modulation of cellular senescence: Implications for breast cancer progression and therapeutic strategies. Eur J Pharmacol 2025; 997:177475. [PMID: 40049574 DOI: 10.1016/j.ejphar.2025.177475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 05/02/2025]
Abstract
Senescence, defined by the cessation of cell proliferation, plays a critical and multifaceted role in breast cancer progression and treatment. Senescent cells produce senescence-associated secretory phenotypes (SASP) comprising inflammatory cytokines, chemokines, and small molecules, which actively shape the tumor microenvironment, influencing cancer development, progression, and metastasis. This review provides a comprehensive analysis of the types and origins of senescent cells in breast cancer, alongside their markers and detection methods. Special focus is placed on pharmacological strategies targeting senescence, including drugs that induce or inhibit senescence, their molecular mechanisms, and their roles in therapeutic outcomes when combined with chemotherapy and radiotherapy. By exploring these pharmacological interventions and their impact on breast cancer treatment, this review underscores the potential of senescence-targeting therapies to revolutionize breast cancer management.
Collapse
Affiliation(s)
- Jialing Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Xianlong Shu
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Zilan Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Jie Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China.
| | - Guo Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China.
| |
Collapse
|
25
|
Jiang Y, Liu J, Pan B, Yu S, Hu C, Li Q, Cheng H, Chen L, Jiang M, Xu D, Wang C, Yan J. Prediction of lung adenocarcinoma prognosis and clinical treatment efficacy by telomere-associated gene risk model. Discov Oncol 2025; 16:1102. [PMID: 40515885 PMCID: PMC12167188 DOI: 10.1007/s12672-025-02977-3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 06/11/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND The most prevalent cause of cancer-related death in China and across the globe is lung adenocarcinoma (LUAD). Telomere shortening (TS) has been found to contribute to the development of LUAD. Therefore, our aim is to investigate the impact of telomere-related genes (TRGs) on immunotherapy and clinical prognosis prediction in LUAD. MATERIALS AND METHODS TRGs were obtained from TelNet, while RNA-seq and clinical information were retrieved from the GEO and TCGA databases. TelNet preserves a series of genes known to be engaged in telomere maintenance and also provides information on the type of telomere maintenance mechanism in which the gene is involved. Data pertinent to RNA sequencing and clinical parameters were accessed from two widely-accessed electronic repositories- the GEO and TCGA databases, respectively. We conducted univariate Cox regression analysis in order to recognize prognostic TRGs and employed multivariate Cox regression analysis to develop a risk model for these TRGs. The patients were stratified into high-risk and low-risk groups based on the first quartile of the risk score. The predictive ability and stability of the model were subsequently verified through Kaplan-Meier analysis, ROC curve, and C-index. We investigated the immune landscapes of different risk groups and predicted their responses to immunotherapy. Lastly, we evaluated the sensitivity of different groups to commonly used chemotherapeutic and targeted drugs through drug sensitivity analysis. RESULTS Univariate Cox analysis identified 12 prognostic TRGs, while a signature consisting of 4 prognostic TRGs was constructed through multivariate Cox analysis. Survival analysis indicated a significantly shorter survival time in the high-risk group. The predictive immunotherapy analysis suggested that patients in the high-risk group may have a more favorable response to immunotherapy. Finally, we identified 28 appropriate chemotherapeutic and 51 targeted drugs for different patient groups. CONCLUSION The study has successfully developed a prognostic model for LUAD prediction that takes into account TRGs and predicts both prognosis and response to immunotherapy.
Collapse
Affiliation(s)
- Yan Jiang
- Department of Respiratory and Critical Care Medicine, Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jun Liu
- Department of Cardiovascular, Luzhou Longmatan District People's Hospital, Luzhou, 646000, Sichuan, China
| | - Bi Pan
- Department of Respiratory and Critical Care Medicine, Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Siping Yu
- Department of Pain Management, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chunyan Hu
- Department of Respiratory and Critical Care Medicine, Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Qiancheng Li
- Department of Respiratory and Critical Care Medicine, Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Hong Cheng
- Department of Respiratory and Critical Care Medicine, Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Ling Chen
- Department of Respiratory and Critical Care Medicine, Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Min Jiang
- Department of Respiratory and Critical Care Medicine, Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Die Xu
- Department of Respiratory and Critical Care Medicine, Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chuanhui Wang
- Department of Respiratory and Critical Care Medicine, Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Jie Yan
- Department of Respiratory and Critical Care Medicine, Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
26
|
Zhang X, Su GH, Bao TS, He WP, Wang YY, Zhou YQ, Xie JX, Wang F, Lu R, Zhang S, Yi SQ, Li Q, Jiang SH, Li H, Hu LPP, Li J, Xu J. TNS4 promotes lymph node metastasis of gastric cancer by interacting with integrin Β1 and inducing the activation of fibroblastic reticular cell. Cancer Cell Int 2025; 25:204. [PMID: 40481538 PMCID: PMC12144749 DOI: 10.1186/s12935-025-03830-x] [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: 10/14/2024] [Accepted: 05/15/2025] [Indexed: 06/11/2025] Open
Abstract
Lymph node (LN) metastasis of gastric cancer (GC) is one of the important pathways of GC metastasis, indicating the clinical staging and prognosis of patients. To investigate the underlying mechanism during the process of GC-induced LN metastasis, 7 pairs of GC tissues, paracancerous (PC) tissues, GC-positive LN (LN.P) and GC-negative LN (LN.N) tissues from GC patients with homogeneity were selected for RNA sequencing (RNA-seq) analysis. Tensin 4 (TNS4) was screened out and found to be significantly upregulated in LN.P tissues and closely related with the characteristics of GC. In vitro and in vivo experiments demonstrated that knockdown of TNS4 could significantly inhibit LN metastasis of GC cells and activation of fibroblastic reticular cells (FRCs) in LNs, thus inhibiting LN expansion induced by tumor cell invasion. Moreover, TNS4 was found to be interacted with integrin beta 1 (ITGB1) on FRCs, thereby affecting the binding of transforming growth factor β1 (TGF-β1) to ITGB1 and subsequently regulating downstream signaling molecules, and supporting the GC cell-induced LN metastasis.
Collapse
Affiliation(s)
- Xiang Zhang
- State Key Laboratory of Systems Medicine for Cancer, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Guang-Hong Su
- State Key Laboratory of Systems Medicine for Cancer, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Tian-Shang Bao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Pai He
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yang-Yang Wang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yao-Qi Zhou
- State Key Laboratory of Systems Medicine for Cancer, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Jia-Xuan Xie
- State Key Laboratory of Systems Medicine for Cancer, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Fei Wang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Rui Lu
- State Key Laboratory of Systems Medicine for Cancer, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Shan Zhang
- State Key Laboratory of Systems Medicine for Cancer, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Shuang-Qin Yi
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Hachioji, Japan
| | - Qing Li
- State Key Laboratory of Systems Medicine for Cancer, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Shu-Heng Jiang
- State Key Laboratory of Systems Medicine for Cancer, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Li
- State Key Laboratory of Systems Medicine for Cancer, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China.
| | - Li-Peng P Hu
- State Key Laboratory of Systems Medicine for Cancer, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China.
| | - Jun Li
- State Key Laboratory of Systems Medicine for Cancer, Ren Ji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China.
| | - Jia Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
27
|
He J, Akhtar A, Li J, Wei Q, Yuan Y, Ran J, Ma Y, Chen D. Integration of Bulk and Single-Cell Transcriptomics Reveals BCL2L14 as a Novel IGKC+ T Cell-Associated Therapeutic Target in Breast Cancer. J Inflamm Res 2025; 18:7215-7234. [PMID: 40491783 PMCID: PMC12147929 DOI: 10.2147/jir.s523147] [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: 03/06/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025] Open
Abstract
Background The tumor microenvironment and biomarkers play a pivotal role in breast cancer research, yet there remains a pressing need for effective biomarkers. This study focuses on identifying a novel IGKC+ T Cell subpopulation and its related biomarkers to pave the way for innovative targeted therapies and improved clinical outcomes. Methods We first performed single-cell RNA sequencing (scRNA-seq) analysis to characterize immune cell heterogeneity within the tumor microenvironment, leading to the identification of series cell subpopulation. Then, by performing univariate analysis to correlate cell proportions with patient prognosis, we identified a novel IGKC+ T cell subpopulation. Next, we applied bulk RNA-seq deconvolution algorithms to estimate the abundance of this subpopulation across breast cancer cohorts. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were employed to identify genes associated with the IGKC+ T cell population. To pinpoint key regulatory genes, we applied machine learning algorithms. Based on the hub genes identified, we constructed a prognostic risk model and developed a nomogram to aid clinical decision-making. Immune infiltration patterns were further assessed in high- vs low-risk groups defined by the model. Finally, functional validation was performed through overexpression of BCL2L14 in vitro, and downstream signaling pathways were examined. Results We identified the novel IGKC+ T cell subpopulation and core genes. Machine learning pinpointed BCL2L14, IGHD, MAPT-AS1, NT5DC4, and TNIP3 as key regulators of breast cancer progression in this subpopulation. The model stratified patients into high- and low-risk groups, with high-risk patients showing worse prognosis and weaker immune infiltration. Overexpression of BCL2L14 was experimentally demonstrated to accelerate breast cancer progression, linked to enhanced phosphorylation of the NF-κB pathway. Conclusion Our results underscore BCL2L14 as a potential driver within the novel T-cell subpopulation and a critical biomarker for breast cancer diagnosis. These findings provide a basis for developing advanced diagnostic tools and targeted therapies, which may ultimately enhance patient prognosis.
Collapse
Affiliation(s)
- Jiaming He
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Aiman Akhtar
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jing Li
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Qiang Wei
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yang Yuan
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jianhua Ran
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yongping Ma
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Dilong Chen
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Three Gorges Medical College, Chongqing, 404120, People’s Republic of China
- NMPA Key Laboratory for Quality Monitoring of Narcotic Drugs and Psychotropic Substances, Chongqing Institute for Food and Drug Control, Chongqing, 401120, People’s Republic of China
| |
Collapse
|
28
|
Yuan JM, Kensler TW, Dacic S, Hartman DJ, Wang R, Balogh PA, Sufka P, Turner MA, Fuhrer K, Seigh L, Pham YTH, Adams-Haduch J, Valacchi G, Singh SV, Herman JG, Wilson DO. Randomized Phase II Clinical Trial of Sulforaphane in Former Smokers at High Risk for Lung Cancer. Cancer Prev Res (Phila) 2025; 18:335-345. [PMID: 40041932 DOI: 10.1158/1940-6207.capr-24-0386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/27/2024] [Accepted: 03/03/2025] [Indexed: 06/18/2025]
Abstract
Experimental studies have shown that dietary isothiocyanates reduced cellular proliferative marker Ki-67 and increased apoptotic markers caspase-3 and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) in animals, but human data are lacking. The present study was to assess whether sulforaphane would stop/reverse the progression of bronchial histopathology, reduce the Ki-67 index, and/or increase caspase-3 and TUNEL indices in humans. A randomized clinical trial (NCT03232138) was conducted in former smokers. Forty-three subjects were randomly assigned to the placebo or the treatment with a potential daily dose of 95 μmol sulforaphane for 12 months. The endpoints were the changes in histopathology scores and Ki-67, caspase-3, and TUNEL indices in post- versus pretreatment bronchial biopsies. Thirty-seven participants (17 in the sulforaphane and 20 in the placebo group) completed the study. Supplementation of sulforaphane did not show significant impact on bronchial histopathology but significantly reduced the Ki-67 index with a 20% decrease in the sulforaphane group and a 65% increase in the placebo (P = 0.014). The difference was even greater in high-density (3+) positive Ki-67, with a 44% decrease in the sulforaphane group compared with a 71% increase in the placebo (P = 0.004). Higher bioavailability of sulforaphane was correlated with greater reduction of the Ki-67 index (P for trend = 0.019). Sulforaphane treatment had no impact on the caspase-3 or TUNEL index in bronchial biopsies. No severe adverse event was observed in the study participants. The findings of oral sulforaphane that significantly reduced the Ki-67 index in bronchial tissue support further development as a potential chemopreventive agent against lung cancer development. Prevention Relevance: High intake of cruciferous vegetables and their sulforaphane is associated with lower incidence of lung cancer in humans and animal models. This clinical trial has demonstrated that oral supplementation of sulforaphane for 12 months significantly reduced the Ki-67 index, a potential surrogate endpoint of biomarkers for lung cancer risk.
Collapse
Affiliation(s)
- Jian-Min Yuan
- Cancer Epidemiology and Prevention Program, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Thomas W Kensler
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sanja Dacic
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Douglas J Hartman
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Renwei Wang
- Cancer Epidemiology and Prevention Program, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Paula A Balogh
- Cancer Epidemiology and Prevention Program, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Pamela Sufka
- Cancer Epidemiology and Prevention Program, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Melissa A Turner
- Cancer Epidemiology and Prevention Program, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Kimberly Fuhrer
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lindsey Seigh
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yen Thi-Hai Pham
- Cancer Epidemiology and Prevention Program, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jennifer Adams-Haduch
- Cancer Epidemiology and Prevention Program, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Giuseppe Valacchi
- Department of Animal Science, Plants for Human Health Institute, North Carolina State University, Kannapolis, North Carolina
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy
- Department of Food and Nutrition, Kyung Hee University, Seoul, South Korea
| | - Shivendra V Singh
- Cancer Epidemiology and Prevention Program, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - James G Herman
- Cancer Epidemiology and Prevention Program, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Division of Hematology & Oncology, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David O Wilson
- Cancer Epidemiology and Prevention Program, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
29
|
Wang X, Zhou D, Jin C, Shi Z, Wen D, Bi L. Establishment of patient-derived xenograft models in Chinese patients with multiple myeloma: Insights into therapeutic responsiveness and molecular subtyping. Transl Oncol 2025; 56:102385. [PMID: 40203791 PMCID: PMC12011171 DOI: 10.1016/j.tranon.2025.102385] [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: 11/05/2024] [Revised: 03/31/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025] Open
Abstract
Multiple myeloma (MM), a malignant hematologic tumor characterized by the proliferation of monoclonal plasma cells, remains incurable with high relapse rates despite advances in treatment. Patient-derived xenograft (PDX) models have emerged as a promising tool for understanding MM's complex pathophysiology and testing therapeutic responses. In this study, we successfully developed PDX models from three patients with MM by subcutaneously engrafting their tumor cells into immunodeficient NCG mice. These models accurately mirrored the clinical drug responses of their corresponding patient cases, exhibiting similar drug sensitivities and resistance patterns. Omics profiling facilitated the alignment of PDX models with specific molecular subgroups identified in current MM research, enhancing the models' clinical relevance. The concordance between PDX models and clinical data confirms the utility of these models in simulating patient-specific responses and advancing personalized treatment strategies. This study validates the effectiveness of PDX models established by subcutaneous engraftment of tumor cells in replicating human disease and treatment responses, thus providing a robust platform for future personalized treatments and development of targeted interventions in Chinese MM patients.
Collapse
Affiliation(s)
- Xueju Wang
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, PR China
| | - Di Zhou
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, PR China
| | - Chanjuan Jin
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, PR China
| | - Zhangzhen Shi
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, PR China
| | - Danyi Wen
- Shanghai LIDE Biotech, Co. Ltd., Pudong, Shanghai, 200120, PR China
| | - Lintao Bi
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, PR China.
| |
Collapse
|
30
|
Li Y, Liu W, Liu C, Wang G, Zhou X. LncRNA SNHG25 facilitates colorectal cancer progression by upregulating PPP2R2D expression through sponging miR-329-3p. Cytotechnology 2025; 77:89. [PMID: 40256259 PMCID: PMC12008101 DOI: 10.1007/s10616-025-00753-3] [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: 01/10/2025] [Accepted: 04/08/2025] [Indexed: 04/22/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) have been evidenced to function as pivotal modulators in tumorigenesis. LncRNA SNHG25 is highly expressed in colorectal cancer (CRC), but its specific function in CRC has not been elucidated yet. The expression of SNHG25, miR-329-3p, and PPP2R2D was determined using qRT-PCR analysis and western blot analysis. The influence of the SNHG25/miR-329-3p/PPP2R2D axis on CRC progression was explored through in vitro assays including CCK-8, colony formation, wound healing, Transwell assays and in vivo orthotopic xenografts assay. The interaction between miR-329-3p and SNHG25 or PPP2R2D was examined by RNA pull-down, RIP, and luciferase reporter assays. SNHG25 presented high expression in CRC cell lines. Silencing of SNHG25 suppressed the malignant phenotypes of CRC cells in vitro and tumor growth in vivo. MiR-329-3p, which displayed low expression in CRC cells, was sponged by SNHG25. Downregulation of miR-329-3p reversed the inhibitory effects of SNHG25 silencing on CRC cell malignant behaviors. Additionally, PPP2R2D served as a miR-329-3p downstream target, whose expression was downregulated by overexpressing miR-329-3p. Importantly, overexpression of PPP2R2D rescued SNHG25 silencing-induced repression on CRC cell malignancy. SNHG25 plays a carcinogenic role in CRC via regulation of the miR-329-3p/PPP2R2D axis. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-025-00753-3.
Collapse
Affiliation(s)
- Yuanqiang Li
- Department of Gastrointestinal Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People’s Hospital, No. 183 Yiling Road, Wujiagang District, Yichang City, Hubei Province China
| | - Weipeng Liu
- Department of Gastrointestinal Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People’s Hospital, No. 183 Yiling Road, Wujiagang District, Yichang City, Hubei Province China
| | - Chao Liu
- Department of Gastrointestinal Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People’s Hospital, No. 183 Yiling Road, Wujiagang District, Yichang City, Hubei Province China
| | - Guangsheng Wang
- Department of Gastrointestinal Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People’s Hospital, No. 183 Yiling Road, Wujiagang District, Yichang City, Hubei Province China
| | - Xin Zhou
- Department of Gastrointestinal Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
- Yichang Central People’s Hospital, No. 183 Yiling Road, Wujiagang District, Yichang City, Hubei Province China
| |
Collapse
|
31
|
Wang T, Jia N, Gao S, Liu S, Liu M, Zhang H. Engrailed-2 (EN2) protein in cervical mucus: a novel biomarker for endometrial carcinoma. Clin Transl Oncol 2025; 27:2484-2493. [PMID: 39607581 DOI: 10.1007/s12094-024-03799-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024]
Abstract
OBJECTIVE This study aims to demonstrate that the EN2 protein in cervical mucus may serve as a novel biomarker for screening endometrial cancer. MATERIALS AND METHODS This study included 133 patients who were treated at Beijing Obstetrics and Gynecology Hospital. According to the pathological results of hysteroscopy endometrial biopsy, the patients were divided into endometrial cancer group (n = 55), endometrial atypical hyperplasia group (n = 16), benign lesion group (n = 28), and control group (n = 34). All patients collected cervical mucus before hysteroscopy, and the level of EN2 protein in cervical mucus was detected by ELISA in the four groups of patients. Nine patients who underwent surgical treatment for endometrial cancer were included, and endometrial cancer lesions and adjacent tissues without endometrial cancer infiltration were taken from the endometrial cancer lesions and cervical tissues. Fifteen patients who underwent hysterectomy for benign lesions were included, and endometrial and cervical tissues were taken from the endometrial cancer lesions and adjacent tissues without endometrial cancer infiltration. PT-PCR, immunohistochemistry, and WB were used to verify the expression differences of EN2 protein in cancerous lesions and endometrial tissues without endometrial infiltration, cervical tissues, and endometrial tissues without endometrial lesions. In addition, HEC1B, KLE, and HeLa cell lines were used to characterize EN2 overexpression in endometrial cancer cells. Immunohistochemistry, RT-PCR, and confocal analysis were used to detect the expression of EN2 protein in different cell lines. RESULTS In different groups, the average concentration of EN2 protein in cervical mucus in the EC group was significantly higher than that in the benign group and the normal control group (573.9 ± 123.4 ng/mL vs 153.5 ± 106.2 ng/mL and 153.0 ± 107.5 ng/mL, P < 0.001). The concentration of EN2 protein in EIN3 case specimens was significantly higher than that in the normal control group (P < 0.001). ROC analysis showed that the optimal threshold for diagnosing endometrial cancer in cervical mucus was 321.1 ng/ml, with a sensitivity of 92.6% and specificity of 95.4% for distinguishing EINIII, EC, and non-cancerous cases. In clinical specimens, the expression level of EN2 protein in endometrial cancer tissues was significantly higher than that in endometrial tissues and cervical tissues without endometrial cancer infiltration. In addition, PT-PCR, immunohistochemistry, suggest that EN2 protein is overexpressed in endometrial cancer cell lines compared to cervical adenocarcinoma cells (P < 0.01). CONCLUSION The concentration of EN2 protein in cervical mucus can effectively identify endometrial cancer and precancerous lesions. The increased expression of EN2 protein in endometrial cancer lesions leads to an increase in the concentration of EN2 protein in the cervical mucus of endometrial cancer patients.This is a small single-center study, and larger studies are needed to determine the role of EN2 protein in the diagnosis of endometrial cancer.
Collapse
Affiliation(s)
- Tong Wang
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No.17 QiHeLou Street, Dongcheng District, Beijing, 100006, China.
| | - Ningyi Jia
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No.17 QiHeLou Street, Dongcheng District, Beijing, 100006, China
| | - Songkun Gao
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, No.17 QiHeLou Street, Dongcheng District, Beijing, 100006, China
| | - Shengjie Liu
- Department of Urology, Beijing Hospital, No. 1 DaHua Road, Dongcheng District, Beijing, China
- National Center of Gerontology, No. 1 DaHua Road, Dongcheng District, Beijing, China
| | - Ming Liu
- Department of Urology, Beijing Hospital, No. 1 DaHua Road, Dongcheng District, Beijing, China
- National Center of Gerontology, No. 1 DaHua Road, Dongcheng District, Beijing, China
| | - Hong Zhang
- Institute of Cardiovascular Sciences, Peking University Health Science Center, No. 38 Xueyuan Road, Handian District, Beijing, China.
| |
Collapse
|
32
|
Morparia S, Metha C, Suvarna V. Recent advancements of betulinic acid-based drug delivery systems for cancer therapy (2002-2023). Nat Prod Res 2025; 39:3260-3280. [PMID: 39385745 DOI: 10.1080/14786419.2024.2412838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 09/14/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
Betulinic acid, a compound classified as a pentacyclic triterpenoid, is found in abundance in a variety of medicinal plants and natural substances. Its broad spectrum of biological and medicinal properties, particularly its potent antitumor activity, has gained significant attention in recent years. The anticancer properties of betulinic acid are governed by mitochondrial signalling pathways and it exhibit selectivity for cancerous tissue, leaving non-cancerous cells and normal tissue unharmed. This characteristic is particularly valuable in chemo-resistant cases. Nevertheless, the medicinal potential of betulinic acid is hindered by its poor water solubility and short half-life, leading to sub-optimal effectiveness. This issue is being tackled by a variety of nano-sized drug delivery systems, such as polymeric nanoparticles, magnetic nanoparticles, polymeric conjugates, nanoemulsions, liposomes, nanosuspensions, carbon nanotubes, and cyclodextrin complexes. This article focuses on recent advances in nanoformulations that are tailored to the delivery of betulinic acid with enhanced effectiveness.
Collapse
Affiliation(s)
- Saurabh Morparia
- Department of Pharmaceutical Analysis & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, Maharashtra, India
| | - Chaitanya Metha
- Department of Pharmaceutical Analysis & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, Maharashtra, India
| | - Vasanti Suvarna
- Department of Pharmaceutical Analysis & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, Maharashtra, India
| |
Collapse
|
33
|
Liu H, Xia H, Yin X, Qin A, Zhang W, Feng S, Jin J. Study on the Differentiation of Infiltrating Breast Cancer Molecular Subtypes Based on Ultrasound Radiomics. Clin Breast Cancer 2025; 25:e450-e460. [PMID: 40044534 DOI: 10.1016/j.clbc.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/05/2025] [Accepted: 01/15/2025] [Indexed: 05/25/2025]
Abstract
OBJECTIVE To establish and validate a 2-dimensional ultrasound (US) radiomics model for the noninvasive preoperative differentiation of various molecular subtypes of infiltrating breast cancer (IBC). METHODS A retrospective analysis of 210 female patients diagnosed with IBC through surgical operation or needle biopsy pathology at our hospital between May 2019 and February 2024 was conducted. Relevant data were collected to establish predictive models for different molecular subtypes of IBC. RESULTS Based on 5936 US radiomics features, 39, 25 and 19 optimal features were identified for the differentiation of luminal versus nonluminal types, luminal A versus luminal B types and human epidermal growth factor receptor 2 (HER2) overexpression versus triple-negative (TN) IBC subgroups, respectively. The corresponding areas under the curve for the training and validation sets were 0.901 and 0.752 (luminal vs. nonluminal), 0.931 and 0.773 (luminal A vs. luminal B) and 0.962 and 0.842 (HER2 overexpression vs. TN), respectively, indicating robust discriminatory performance of these models for different pathological molecular subtypes of IBC. CONCLUSION A radiomics model based on US images is capable of effectively differentiating between various molecular subtypes of IBC prior to surgery, holding promise in assisting medical professionals in crafting tailored diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hanqin Liu
- Department of Ultrasound, Medical Imaging Center, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225100, China
| | - Han Xia
- Department of Ultrasound, Medical Imaging Center, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225100, China
| | - Xiaoxiao Yin
- Department of Ultrasound, Medical Imaging Center, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225100, China
| | - Aiping Qin
- Department of Ultrasound, Medical Imaging Center, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225100, China
| | - Wen Zhang
- Department of Ultrasound, Medical Imaging Center, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225100, China
| | - Shuang Feng
- Department of Ultrasound, Medical Imaging Center, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225100, China
| | - Jing Jin
- Department of Ultrasound, Medical Imaging Center, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225100, China.
| |
Collapse
|
34
|
Cobbs C, Chesnut GT, Shafi AA. Understanding Racial Disparities in Prostate Cancer: A Multifaceted Approach. Cancer Med 2025; 14:e70979. [PMID: 40444484 PMCID: PMC12123386 DOI: 10.1002/cam4.70979] [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: 01/14/2025] [Revised: 05/05/2025] [Accepted: 05/12/2025] [Indexed: 06/02/2025] Open
Abstract
Prostate cancer (PCa) remains a significant public health challenge in the United States, disproportionately affecting African American (AA) men, who face higher incidence rates, more aggressive disease, and elevated mortality compared to Caucasian American (CA) men. This review explores the multifactorial underpinnings of these disparities, integrating genomic, socioeconomic, environmental, and systemic contributors. Genomic analyses reveal that AA men harbor distinct molecular alterations, including higher frequencies of FOXA1, BRAF, and CHD1 mutations, as well as DNA damage repair defects, highlighting the critical need for population-specific precision medicine. Immune-oncologic pathways and stromal interactions within the tumor microenvironment further underscore biological differences driving aggressive disease phenotypes. Concurrently, adverse social determinants-including limited access to care, lower PSA screening rates, delayed treatment, medical mistrust, and underrepresentation in clinical trials-contribute to poorer outcomes. Despite these challenges, evidence from equal-access healthcare systems indicates that when provided equitable treatment, AA men can achieve outcomes comparable to or better than their CA counterparts. This review emphasizes actionable strategies to reduce disparities, including increasing AA representation in clinical trials, enhancing culturally competent patient-provider communication, improving access to early detection and high-quality care, and expanding community-based outreach initiatives. A holistic, interdisciplinary approach is essential to dismantle systemic barriers and achieve health equity in prostate cancer outcomes.
Collapse
Affiliation(s)
- Charles Cobbs
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of SurgeryUniformed Services University of the Health SciencesBethesdaMarylandUSA
| | - Gregory T. Chesnut
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of SurgeryUniformed Services University of the Health SciencesBethesdaMarylandUSA
- Urology Service, Department of SurgeryWalter Reed National Military Medical CenterBethesdaMarylandUSA
| | - Ayesha A. Shafi
- Center for Prostate Disease Research, Murtha Cancer Center Research Program, Department of SurgeryUniformed Services University of the Health SciencesBethesdaMarylandUSA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc.BethesdaMarylandUSA
| |
Collapse
|
35
|
Li BR, Wang T, Hu HF, Wu D, Zhou CJ, Ji SR, Zhuo QF, Li Z, Wang ZL, Fan GX, Jing DS, Yu CY, Qin Y, Chen XM, Xu JF, Xu XW. Acyl-CoA thioesterase 8 induces gemcitabine resistance via regulation of lipid metabolism and antiferroptotic activity in pancreatic ductal adenocarcinoma. Acta Pharmacol Sin 2025; 46:1742-1756. [PMID: 39939803 PMCID: PMC12098905 DOI: 10.1038/s41401-025-01477-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/08/2025] [Indexed: 02/14/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) comprises a group of highly malignant tumors of the pancreas. Metabolic reprogramming in tumors plays a pivotal role in promoting cancer progression. However, little is known about the metabolic alterations in tumors that drive cancer drug resistance in patients with PDAC. Here, we identified acyl-CoA thioesterase 8 (ACOT8) as a key player in driving PDAC gemcitabine (GEM) resistance. The expression of ACOT8 is significantly upregulated in GEM-resistant PDAC tissues and is closely associated with poor survival in patients with PDAC. Gain- and loss-of-function studies have shown that ACOT8 drives PDAC GEM resistance both in vitro and in vivo. Mechanistically, ACOT8 regulates cellular cholesterol ester (CE) levels, decreases the levels of phosphatidylethanolamines (PEs) that bind to polyunsaturated fatty acids and promote peroxisome activation. The knockdown of ACOT8 promotes ferroptosis and increases the chemosensitivity of tumors to GEM by inducing ferroptosis-associated pathway activation in PDAC cell lines. The combination of orlistat, an ACOT8 inhibitor, and GEM significantly inhibited tumor growth in PDAC organoid and mouse models. This study reveals the biological importance of ACOT8 and provides a potential combination therapy for treating patients with advanced GEM-resistant PDAC.
Collapse
Affiliation(s)
- Bo-Rui Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Hepatopancreatobiliary Surgery, First College of Clinical Medical Science, Three Gorges University, Yichang, 443003, China
- People's Hospital of China Three Gorges University, Yichang, 443099, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Ting Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Hai-Feng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
- Department of General Surgery, First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Di Wu
- Department of Hepatopancreatobiliary, Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Chen-Jie Zhou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Shun-Rong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Qi-Feng Zhuo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zheng Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zhi-Liang Wang
- Department of Hepatopancreatobiliary, Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Gui-Xiong Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - De-Sheng Jing
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chong-Yuan Yu
- Department of Hepatopancreatobiliary, Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xue-Min Chen
- Department of Hepatopancreatobiliary, Third Affiliated Hospital of Soochow University, Changzhou, 213000, China.
| | - Jun-Feng Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xiao-Wu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
36
|
Gou M, Zhang H, Qian N, Zhang Y, Sun Z, Li G, Wang Z, Dai G. Deep learning radiomics analysis for prediction of survival in patients with unresectable gastric cancer receiving immunotherapy. Eur J Radiol Open 2025; 14:100626. [PMID: 39807092 PMCID: PMC11728962 DOI: 10.1016/j.ejro.2024.100626] [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: 10/28/2024] [Revised: 12/03/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
Objective Immunotherapy has become an option for the first-line therapy of advanced gastric cancer (GC), with improved survival. Our study aimed to investigate unresectable GC from an imaging perspective combined with clinicopathological variables to identify patients who were most likely to benefit from immunotherapy. Method Patients with unresectable GC who were consecutively treated with immunotherapy at two different medical centers of Chinese PLA General Hospital were included and divided into the training and validation cohorts, respectively. A deep learning neural network, using a multimodal ensemble approach based on CT imaging data before immunotherapy, was trained in the training cohort to predict survival, and an internal validation cohort was constructed to select the optimal ensemble model. Data from another cohort were used for external validation. The area under the receiver operating characteristic curve was analyzed to evaluate performance in predicting survival. Detailed clinicopathological data and peripheral blood prior to immunotherapy were collected for each patient. Univariate and multivariable logistic regression analysis of imaging models and clinicopathological variables was also applied to identify the independent predictors of survival. A nomogram based on multivariable logistic regression was constructed. Result A total of 79 GC patients in the training cohort and 97 patients in the external validation cohort were enrolled in this study. A multi-model ensemble approach was applied to train a model to predict the 1-year survival of GC patients. Compared to individual models, the ensemble model showed improvement in performance metrics in both the internal and external validation cohorts. There was a significant difference in overall survival (OS) among patients with different imaging models based on the optimum cutoff score of 0.5 (HR = 0.20, 95 % CI: 0.10-0.37, P < 0.001). Multivariate Cox regression analysis revealed that the imaging models, PD-L1 expression, and lung immune prognostic index were independent prognostic factors for OS. We combined these variables and built a nomogram. The calibration curves showed that the C-index of the nomogram was 0.85 and 0.78 in the training and validation cohorts. Conclusion The deep learning model in combination with several clinical factors showed predictive value for survival in patients with unresectable GC receiving immunotherapy.
Collapse
Affiliation(s)
- Miaomiao Gou
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Hongtao Zhang
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Niansong Qian
- Department of Thoracic Oncology, The Eighth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Yong Zhang
- Department of Medical Oncology, The Second Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Zeyu Sun
- R&D Center, Keya Medical Technology Co., Ltd, Beijing, PR China
| | - Guang Li
- R&D Center, Keya Medical Technology Co., Ltd, Beijing, PR China
| | - Zhikuan Wang
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Guanghai Dai
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| |
Collapse
|
37
|
Bock K, Peltzer J, Liu W, Colgrove Y, Smirnova I, Siengsukon C. Sleep quality and lymphedema in breast cancer survivors: a mixed method analysis. J Cancer Surviv 2025; 19:978-992. [PMID: 38183577 DOI: 10.1007/s11764-023-01516-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 12/17/2023] [Indexed: 01/08/2024]
Abstract
PURPOSE The purpose of this convergent mixed methods study was to assess the perceptions and characteristics of sleep in breast cancer survivors (BCSs) and elucidate perceptions of sleep among BCS with lymphedema. METHODS Participants were BCS with and without lymphedema. Both groups completed the Pittsburgh Sleep Quality Index (PSQI), PROMIS® Sleep Disturbance (8a short form), and wore an actigraph on their wrist to capture sleep/wake cycles for 7 days/nights while logging their sleep using a sleep diary. The coefficient of variation of sleep efficiency was calculated from the sleep diary to assess intraindividual variability. In addition, a subsample of BCS with lymphedema participated in a semi-structured qualitative interview. The qualitative data was analyzed separately, and the themes were applied to provide a more nuanced explanation of the quantitative outcomes. RESULTS The BCS with lymphedema (n=23) had a significant difference in PSQI (p=0.002), PROMIS® Sleep Disturbance (p=0.084), and sleep efficiency coefficient of variation (p=0.014) compared to BCS without lymphedema (n=23). There were no statistically significant differences between groups in the actigraphy results. BCS with lymphedema perceived that lymphedema management contributed to their sleep disturbance, further exacerbating their mind/body fatigue. CONCLUSION This study provides the foundation for future research to investigate the integration of sleep interventions with lymphedema management for holistic survivorship care for BCS with lymphedema. IMPLICATIONS FOR CANCER SURVIVORS An innovative sleep health intervention designed to consider the unique factors contributing to sleep disturbance in BCS with lymphedema will fill a gap in their post-cancer treatment quality of life.
Collapse
Affiliation(s)
- Karen Bock
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Jill Peltzer
- School of Nursing, University of Kansas Medical Center, Kansas City, KS, USA
| | - Wen Liu
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS, USA
| | - Yvonne Colgrove
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS, USA
| | - Irina Smirnova
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS, USA
| | - Catherine Siengsukon
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
38
|
Garcia-Murillas I, Abbott CW, Cutts RJ, Boyle SM, Pugh J, Keough KC, Li B, Pyke RM, Navarro FCP, Chen RO, Dunne K, Bunce C, Johnston SRD, Ring A, Russell S, Evans A, Skene A, Smith IE, Turner NC. Whole genome sequencing-powered ctDNA sequencing for breast cancer detection. Ann Oncol 2025; 36:673-681. [PMID: 39914664 DOI: 10.1016/j.annonc.2025.01.021] [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: 09/15/2024] [Revised: 12/20/2024] [Accepted: 01/28/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Circulating tumour DNA (ctDNA)-based detection of molecular residual disease (MRD) presents a strategy to identify patients at high risk of relapse. In this article, we profile early breast cancer patients with an ultrasensitive, whole genome sequencing (WGS)-based, tumour-informed ctDNA platform. MATERIALS AND METHODS We analysed 617 plasma samples (median 8, range 2-14) from 78 patients (23 triple-negative breast cancer, 35 human epidermal growth factor receptor 2-positive, 18 hormone receptor-positive, and 2 unknown). Samples were collected at diagnosis before therapy, cycle 2 of neoadjuvant chemotherapy, post-surgery after neoad'juvant therapy if administered, every 3 months during the first year, and every 6 months thereafter. Plasma DNA was analysed using the NeXT Personal MRD platform, a tumour-informed WGS approach to produce personalized ctDNA sequencing panels tracking a median of 1451 variants per patient. MRD detection was correlated with clinical outcomes. RESULTS ctDNA was detected at levels ranging from 2.19 parts per million (PPM) to 204 900 PPM (median 405 PPM), with 39% of all ctDNA detections in the ultra-low range <100 PPM. Of patients with samples at diagnosis, 98% (49/50) had ctDNA detected before treatment. At a median follow-up of 76 months (range 5-118 months), detection of ctDNA was associated with high risk of future relapse (P < 0.0001; log-rank test) and shortened overall survival (P < 0.0001) with a median lead time from ctDNA detection to clinical relapse of 15 months (range 0.9-61.5 months). MRD was identified in 100% (11/11) of patients who relapsed, with a median level of ctDNA at first MRD detection of 13.1 PPM. No ctDNA-undetected patients relapsed throughout follow-up (64/64). Comparison with exome-powered MRD detection assays showed improved sensitivity and lead time. CONCLUSIONS A whole genome-powered MRD assay detected breast cancer relapse with a long lead time over clinical relapse, and was strongly associated with relapse-free survival. Rates of ctDNA detection at diagnosis were higher than those reported with exome-based tumour-informed assays.
Collapse
Affiliation(s)
- I Garcia-Murillas
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | | | - R J Cutts
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | | | - J Pugh
- Personalis Inc., Fremont, USA
| | | | - B Li
- Personalis Inc., Fremont, USA
| | | | | | | | - K Dunne
- The Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - C Bunce
- Clinical Trials Unit, Royal Marsden Hospital, London, UK
| | | | - A Ring
- Breast Unit, Royal Marsden Hospital, London, UK
| | - S Russell
- Hinchingbrooke Hospital, Hinchingbrooke Park, Huntingdon, UK
| | - A Evans
- Poole General Hospital, Dorset, UK
| | - A Skene
- Royal Bournemouth Hospital, Bournemouth, UK
| | - I E Smith
- Breast Unit, Royal Marsden Hospital, London, UK
| | - N C Turner
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK; The Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK; Breast Unit, Royal Marsden Hospital, London, UK.
| |
Collapse
|
39
|
Liu X, Shi C, Han B, Yang J. Geographic Distribution of Racial Differences in Renal Cell Carcinoma Mortality. Clin Genitourin Cancer 2025; 23:102324. [PMID: 40157898 DOI: 10.1016/j.clgc.2025.102324] [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: 11/02/2024] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
OBJECTIVE To examine the geographic variations in Renal cell carcinoma (RCC) -specific death disparities from competing causes among Hispanic, non-Hispanic White, non-Hispanic Black, and Asian/Pacific Islander RCC patients. RCC outcomes in specific racial/ethnic population warrants further research and it is unknown whether racial/ethnic differences in RCC survival vary geographically within the US. METHODS This retrospective cohort study was conducted to assess all RCC patients from 2014 to 2021. Data was extracted from the Surveillance, Epidemiology, and End Results (SEER) database. The primary outcome was RCC-related mortality. RESULTS The study included 85,975 patients with RCC from 16 geographic areas within the SEER database. Kaplan-Meier analysis showed that Hispanic patients had the worst survival outcome (P < .001 by log rank test). In the multivariable competing-risks regression, Hispanics had a higher risk of cancer-specific mortality (hazard ratio [HR] 1.29, 95% CI, 1.20-1.38, P ˂ .001) compared with non-Hispanic Whites. The increase in the risk of RCC-related death with Hispanic race/ethnicity was consistent across all major subgroups stratified by the covariables. In stratified analyses of geographic regions, there were 3 areas in which Hispanics had worse RCC-specific survival (Los Angeles: HR 1.22, 95% CI, 1.06-1.41, P = .005; Greater California: HR 1.125, 95% CI, 1.15-1.37, P < .001; Atlanta, Georgia: HR 1.95, 95% CI, 1.32-2.88, P = .001). CONCLUSION These results demonstrate that population-level variations in RCC survival among Hispanics and non-Hispanic Whites were associated with a small number of geographic regions. Targeted interventions in these regions may be conducive to alleviating RCC care differences at the national level.
Collapse
Affiliation(s)
- Xiaoxian Liu
- Department of Nephrology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengqian Shi
- Department of Nephrology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Bin Han
- Department of Nephrology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Yang
- Department of Nephrology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
40
|
Walton WC, Kim SJ. Uncertainty Estimation for Dual View X-ray Mammographic Image Registration Using Deep Ensembles. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025; 38:1829-1845. [PMID: 39313715 DOI: 10.1007/s10278-024-01244-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 07/19/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024]
Abstract
Techniques are developed for generating uncertainty estimates for convolutional neural network (CNN)-based methods for registering the locations of lesions between the craniocaudal (CC) and mediolateral oblique (MLO) mammographic X-ray image views. Multi-view lesion correspondence is an important task that clinicians perform for characterizing lesions during routine mammographic exams. Automated registration tools can aid in this task, yet if the tools also provide confidence estimates, they can be of greater value to clinicians, especially in cases involving dense tissue where lesions may be difficult to see. A set of deep ensemble-based techniques, which leverage a negative log-likelihood (NLL)-based cost function, are implemented for estimating uncertainties. The ensemble architectures involve significant modifications to an existing CNN dual-view lesion registration algorithm. Three architectural designs are evaluated, and different ensemble sizes are compared using various performance metrics. The techniques are tested on synthetic X-ray data, real 2D X-ray data, and slices from real 3D X-ray data. The ensembles generate covariance-based uncertainty ellipses that are correlated with registration accuracy, such that the ellipse sizes can give a clinician an indication of confidence in the mapping between the CC and MLO views. The results also show that the ellipse sizes can aid in improving computer-aided detection (CAD) results by matching CC/MLO lesion detects and reducing false alarms from both views, adding to clinical utility. The uncertainty estimation techniques show promise as a means for aiding clinicians in confidently establishing multi-view lesion correspondence, thereby improving diagnostic capability.
Collapse
Affiliation(s)
- William C Walton
- University of Maryland, Baltimore County, CSEE Department, Baltimore, MD, 21250, USA
- The Johns Hopkins University Applied Physics Laboratory, Laurel, MD, 20723, USA
| | - Seung-Jun Kim
- University of Maryland, Baltimore County, CSEE Department, Baltimore, MD, 21250, USA.
| |
Collapse
|
41
|
Hai ZX, Zhao JN, Liu XR, Qu SP, Lv Q, Wang CY. Effects of Planned Stoma Before Neoadjuvant Chemoradiation in Patients With Endoscopically Obstructing Colorectal Cancer. Am Surg 2025; 91:978-983. [PMID: 40114325 DOI: 10.1177/00031348251329482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
PurposeIn order to investigate whether colorectal cancer (CRC) patients with endoscopic obstruction benefited from a planned stoma before neoadjuvant chemoradiation (nCRT).MethodsPatients who were diagnosed with CRC with endoscopic obstruction at a single clinical center from January 2017 to April 2022 were retrospectively collected. Baseline characteristics and short-term and long-term outcomes were compared between the stoma group and the no stoma group. Statistical analysis was performed using SPSS (version 22.0) software.ResultsA total of 51 CRC patients with endoscopic obstruction were included in this study. Eleven (21.6%) patients received a planned stoma before nCRT, and 40 (78.4%) patients were treated with immediate nCRT. The mean time from diagnosis to nCRT was 30.6 days for the stoma group and 11.9 days for the no stoma group. There was a significant delay in the initiation of nCRT in the stoma group (P < 0.05). In terms of complications, there was a statistical difference between the stoma group and the no stoma group (P < 0.05). Planned stoma before nCRT did not affect survival for patients with endoscopically obstructing CRC (P > 0.05).ConclusionA planned stoma caused delay in nCRT; the no stoma group was more likely to develop perforation or obstruction of the tumor during nCRT. A comprehensive assessment might be needed to determine whether a planned stoma was necessary in CRC patients with endoscopic obstruction.
Collapse
Affiliation(s)
- Zhan-Xiang Hai
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun-Nan Zhao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xu-Rui Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shu-Pei Qu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Quan Lv
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chun-Yi Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
42
|
Liu F, Howard CB, Huda P, Fletcher NL, Bell CA, Blakey I, Agrez M, Thurecht KJ. Immune-modulating nanomedicines for enhanced drug delivery to non-small-cell lung cancer. Biomaterials 2025; 317:123089. [PMID: 39793167 DOI: 10.1016/j.biomaterials.2025.123089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
Immune-modulating peptides have shown potential as novel immune-stimulating agents which enhance the secretion of anticancer cytokines in vitro. However, fast clearance from blood hampers the ability of such peptides to accumulate in the tumour and results in limited therapeutic efficacy in animal studies. To address the fast blood clearance, this work reports the development and validation of a novel polymeric nanoparticle delivery system for the efficient localization of an immunomodulating peptide in the tumour microenvironment (TME). To identify the optimal polymeric nanoparticle for this study, two types of nanoparticles were developed as either branched polymers or micelles that have similar chemical functionality but different sizes. The effect of targeting the nanomedicine to the tumour-specific antigen, glycoprotein GPC-1, was explored using a bispecific antibody (BsAb) that shows an affinity for the cell protein (GPC-1) and the nanoparticle. These systems were evaluated for targeting efficiency and tumour penetration using tumour spheroids of Lewis Lung Cancer (LLC) cells and it was shown that the targeted system significantly enhanced cell association compared to the untargeted control with minor differences in penetration. The lead micelle-peptide conjugates were identified and using in vivo allograft models they were demonstrated to have high delivery efficiency of the peptide to tumours, prolonged blood circulation, enhanced tumour accumulation and tumour suppression that was associated with immune cell recruitment to the tumour.
Collapse
Affiliation(s)
- Feifei Liu
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Training Centre for Innovation in Biomedical Imaging Technology, University of Queensland, QLD, Australia
| | - Christopher B Howard
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Pie Huda
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Nicholas L Fletcher
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Craig A Bell
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Idriss Blakey
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Michael Agrez
- ARC Training Centre for Innovation in Biomedical Imaging Technology, University of Queensland, QLD, Australia; InterK Peptide Therapeutics Limited, Sydney, Australia
| | - Kristofer J Thurecht
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Training Centre for Innovation in Biomedical Imaging Technology, University of Queensland, QLD, Australia.
| |
Collapse
|
43
|
El-Khatib A, Matos K, Rossoff J, Psihogios A. Social Determinants of 6-Mercaptopurine Adherence Among Patients with Acute Lymphoblastic Leukemia/Lymphoma: A Cross-Sectional Analysis. J Adolesc Young Adult Oncol 2025; 14:270-274. [PMID: 39527123 DOI: 10.1089/jayao.2024.0119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Acute lymphoblastic leukemia requires around 18 months of daily oral chemotherapy called 6-mercaptopurine (6-MP) for treatment. Previous research has included a broad age range and shows adolescent/young adult (AYA) age and minoritized race/ethnicity are associated with lower adherence. Few studies have evaluated how these intersecting identities impact adherence. In a retrospective analysis of an AYA sample, we evaluated the relationships between race/ethnicity, area deprivation index (ADI), and insurance with electronically monitored 6-MP adherence. Results showed minoritized race/ethnicity and ADI correlated with adherence. Findings highlight the need for equitable interventions that enhance adherence and improve outcomes among AYAs.
Collapse
Affiliation(s)
- Ayah El-Khatib
- Division of Hematology, Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Kevin Matos
- Department of Medical Social Sciences, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jenna Rossoff
- Division of Hematology, Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Alexandra Psihogios
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Medical Social Sciences, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
44
|
Zhang L, Yuan J, Yao S, Wen G, An J, Jin H, Tuo B. Role of m5C methylation in digestive system tumors (Review). Mol Med Rep 2025; 31:142. [PMID: 40183387 PMCID: PMC11979572 DOI: 10.3892/mmr.2025.13507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Currently, the incidence of digestive system tumors has been increasing annually, thus becoming a prevalent cause of cancer‑related mortalities. Although significant strides have been made in targeting the molecular mechanisms that underpin the development of these tumors, their treatment and prognosis still pose substantial challenges. This is primarily due to the ambiguity of early diagnostic indicators and the fact that most digestive system tumors are detected at an advanced stage. However, epigenetic modifications are capable of altering the expression of oncogenes and regulating biological processes in cancer. In recent years, the study of methylation in relation to tumor pathogenesis has become a focus of prominent research. Among the various types of methylation, 5‑methylcytosine (m5C) methylation plays a crucial role in the development of digestive system tumors and is anticipated to serve as a novel therapeutic target. However, to date, a comprehensive and systematic review concerning the role of m5C methylation in digestive system tumors is lacking. Consequently, the present study reviewed the role of m5C methylation in digestive system tumors such as esophageal cancer, gastric cancer and hepatocellular carcinoma, with the aim of providing a valuable reference for future research endeavors.
Collapse
Affiliation(s)
- Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jianbo Yuan
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, P.R. China
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Guorong Wen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jiaxing An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
45
|
Hu M, Ji L, Jin L, Shao M. Minimally invasive surgery for cervical cancer. Oncol Lett 2025; 29:281. [PMID: 40242268 PMCID: PMC12001325 DOI: 10.3892/ol.2025.15027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 02/14/2025] [Indexed: 04/18/2025] Open
Abstract
The present study aimed to evaluate the oncologic outcomes of minimally invasive radical hysterectomy with no tumor exposure. Briefly, a multicenter, retrospective analysis was conducted between January 2017 and June 2020 involving 350 women with early-stage cervical cancer. Eligible patients were informed of the potential complications and benefits of abdominal radical hysterectomy and laparoscopic radical hysterectomy. During surgery, the use of a uterine manipulator was avoided. Myoma drill and uterine suture techniques were employed, accompanied by protective measures for vaginal closure of the colpotomy, such as clamps, vaginal cuffs or sutures. Specimens were placed in a collection bag, which was extracted through the vaginal route. Over a median follow-up period of 51 months (range, 30-72 months), five patients were lost to follow-up and three refused treatment following surgery; therefore, a total of 342 women with cervical cancer were followed up to the end of the study. The initial stage, according to the International Federation of Gynecology and Obstetrics 2018 classification system, was identified as IA1 with lymphovascular space invasion in 22 cases (6.29%), IA2 in 36 cases (10.29%), IB1 in 137 cases (39.14%), IB2 in 126 cases (36.00%), IIA1 in 14 cases (4.00%) and IIIC1P in 15 cases (4.29%). Histologically, squamous cell carcinoma was diagnosed in 269 patients (76.86%), adenocarcinoma in 75 patients (21.43%) and adenosquamous carcinoma in six patients (1.71%). Lymphovascular invasion was confirmed in 80 patients (22.86%). Lymph nodes were tumor-free in 335 patients (95.71%). After radical hysterectomy, 53 patients underwent brachytherapy and teletherapy, and 30 received chemotherapy alongside brachytherapy and teletherapy. After a median follow-up time of 51 months (range, 30-72 months), the disease-free and overall survival rates were recorded as 95.71% (335/350) and 98.86% (346/350) respectively. In conclusion, minimally invasive surgery using maneuvers to avoid peritoneal contamination yields good oncologic outcomes for patients with early-stage cervical cancer. The findings from the current retrospective analysis suggest that laparoscopic surgery could present a safe oncological option; however, further validation through randomized trials is essential.
Collapse
Affiliation(s)
- Min Hu
- Department of Gynecology, Jinhua Maternity and Child Health Care Hospital, Jinhua, Zhejiang 321000, P.R. China
| | - Limei Ji
- Department of Gynecology, Jinhua Maternity and Child Health Care Hospital, Jinhua, Zhejiang 321000, P.R. China
- Department of Gynecology, Jinhua Hospital of Zhejiang University, Jinhua, Zhejiang 321000, P.R. China
| | - Lanying Jin
- Department of Gynecology, Jinhua Maternity and Child Health Care Hospital, Jinhua, Zhejiang 321000, P.R. China
- Department of Gynecology, Jinhua Hospital of Zhejiang University, Jinhua, Zhejiang 321000, P.R. China
| | - Mingjun Shao
- Department of Gynecology, Jinhua Maternity and Child Health Care Hospital, Jinhua, Zhejiang 321000, P.R. China
- Department of Gynecology, Jinhua Hospital of Zhejiang University, Jinhua, Zhejiang 321000, P.R. China
| |
Collapse
|
46
|
Jiang J, Shen T, Chen D, Dai Z, Wang X, Meng Q, Yang Z, Zhang D, Guo X, Xu J, Gu J, Wang C. FOXM1, a super enhancer-associated gene, is related to poorer prognosis and gemcitabine resistance in pancreatic cancer. Cell Biochem Biophys 2025; 83:2441-2452. [PMID: 39899193 DOI: 10.1007/s12013-024-01653-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2024] [Indexed: 02/04/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive solid tumor; however, the barrier of chemoresistance has yet to be overcome for longer survival. Aberrant gene expression due to epigenetic modification plays an important role in tumorigenesis and treatment. Super enhancers are epigenetic elements that promote targeted gene transcription and ultimately lead to chemoresistance. This study found that the expression of FOXM1 was higher in PDAC tissues and negatively correlated with prognosis. Through RNA sequencing and chromatin immunoprecipitation-sequencing analyses, FOXM1 was found to be regulated by a BRD4-associated super enhancer, which finally promoted gemcitabine resistance via TGFβ/Smad signaling pathway activation. Both TGFβ/Smad-specific inhibitor LY364947 and the BRD4 inhibitor JQ1 decreased the IC50 value of gemcitabine in vitro. Furthermore, combined gemcitabine and JQ1 therapy could not only enhance the therapeutic effect of gemcitabine but also reverse drug resistance in vivo. In conclusion, the super enhancer-associated gene FOMX1 contributes to gemcitabine resistance and is a promising target in PDAC treatment.
Collapse
MESH Headings
- Humans
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Deoxycytidine/therapeutic use
- Gemcitabine
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/metabolism
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Forkhead Box Protein M1/genetics
- Forkhead Box Protein M1/metabolism
- Cell Line, Tumor
- Prognosis
- Transcription Factors/metabolism
- Transcription Factors/antagonists & inhibitors
- Animals
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/metabolism
- Triazoles/pharmacology
- Triazoles/therapeutic use
- Female
- Mice
- Azepines/pharmacology
- Azepines/therapeutic use
- Male
- Cell Cycle Proteins/metabolism
- Antimetabolites, Antineoplastic/pharmacology
- Antimetabolites, Antineoplastic/therapeutic use
- Enhancer Elements, Genetic
- Gene Expression Regulation, Neoplastic/drug effects
- Mice, Nude
- Signal Transduction/drug effects
- Transforming Growth Factor beta/metabolism
- Super Enhancers
- Bromodomain Containing Proteins
Collapse
Affiliation(s)
- Jian Jiang
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tianci Shen
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dan Chen
- Department of Pathology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zihao Dai
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xuelong Wang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qiang Meng
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Zhuo Yang
- Department of Endoscope, General Hospital of Northern Theater Command, Shenyang, China
| | - Di Zhang
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaoyi Guo
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jianqiang Xu
- School of Life and Pharmaceutical Sciences (LPS) & Panjin Institute of Industrial Technology (PIIT), Dalian University of Technology, Panjin, Liaoning, China
| | - Jiangning Gu
- Department of Endoscope, General Hospital of Northern Theater Command, Shenyang, China.
| | - Changmiao Wang
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
47
|
Aubrey BJ, Blonquist T, McMasters M, Hobbs G, McAfee S, Rosenblatt J, Amrein P, Connolly C, Ramos A, Logan E, Dey B, Spitzer T, Avigan D, Chen YB, Ballen K, El-Jawahri A, Fathi AT, Brunner AM. A phase I clinical trial of lenalidomide combined with bortezomib for acute myeloid leukemia or myelodysplastic syndrome relapsing after allogeneic stem cell transplantation. Leuk Res 2025; 153:107693. [PMID: 40250191 DOI: 10.1016/j.leukres.2025.107693] [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: 11/07/2024] [Revised: 02/25/2025] [Accepted: 04/05/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Allogeneic hematopoietic cell transplantation (HCT) may improve long-term survival in patients with MDS or AML but disease relapse following HCT is common, with limited subsequent treatment options and extremely poor post-relapse outcomes. Lenalidomide and bortezomib are therapies which, in this setting, may exert antiproliferative effects, enhance graft-vs-leukemia immune responses, and potentiate chemotherapeutic drugs. OBJECTIVES We sought to evaluate the safety and preliminary efficacy of bortezomib in combination with high dose lenalidomide in patients with AML or MDS relapsing after HCT. STUDY DESIGN We conducted a phase I, dose-escalation, multi-center study of bortezomib added to high dose lenalidomide in patients with AML or MDS relapsing after HCT. We enrolled adult patients with recurrent or progressive AML or MDS after transplant; patients were required to be off systemic immunosuppression, with no evidence of GVHD, and with adequate organ function. Escalating doses of bortezomib were administered on days 2, 5, 9, and 12 of each cycle, while lenalidomide was given at 50 mg daily on days 1-21 of a 28-day cycle. DLTs were assessed during the first 2 cycles of induction and responses were assessed within this period. After achieving a treatment response, patients could proceed to maintenance dosing. The primary endpoint was toxicity and to establish the maximally tolerated dose (MTD) of the combination, while secondary endpoints included response rate and duration of responses. RESULTS 21 patients were enrolled, with a median age of 66 years (range 23-74). The majority of patients enrolled had AML (19/21). Three patients were enrolled at each bortezomib dose level of 0.7 mg/m2, 1.0 mg/m2 and 1.3 mg/m2. One patient experienced dose-limiting toxicity at 1.3 mg/m2 (received <50 % doses) and the cohort was expanded to 6 total patients, establishing this as the recommended phase two dose (RP2D); a maximum tolerated dose (MTD) was not reached. An additional 9 expansion patients were treated at this dose level for a total of 15 patients treated with lenalidomide 50 mg and bortezomib 1.3 mg/m2. During dose expansion, grade 4 toxicities attributed to study drug included grade 4 neutropenia (n = 5), thrombocytopenia (n = 3), and febrile neutropenia (n = 1). Across all cohorts (n = 21), 1 patient (4.8 %) achieved CR and 3 patients (14.3 %) achieved CRi (composite CR/CRi rate of 19 %). Of the 15 patients treated at the RP2D, 4 patients had progressive disease after the first induction cycle, and 2 patients stopped therapy during induction cycle 2. A total of 9 patients completed both induction cycles. Of the 15 patients treated at the RP2D, 1 achieved CR, 2 achieved CRi (composite CR/CRi 20 %, CI 5.7-44.0 %) and 4 patients had stable disease. Chimerism during treatment generally tracked with disease response and one patient with persistent low chimerism also achieved CRi. The single patient achieving CR harbored an NPM1 gene mutation. A total of 3 out of 13 patients tested harbored a TP53 gene mutation and 2 of these patients achieved CRi on study subsequent to receiving DLI before study entry. CONCLUSIONS Bortezomib can be combined with lenalidomide in patients with AML and MDS relapsing after HCT with overall 19 % of patients achieving composite remission. Toxicity was manageable and primarily related to cytopenias during induction. Responding patients included 1 with NPM1-mutated AML as well as 2 harboring TP53 gene mutations, suggesting possible therapeutic benefit in very high-risk disease. Bortezomib and lenalidomide could be safely used following donor lymphocyte infusion without evidence of graft failure or immune-related toxicity.
Collapse
Affiliation(s)
- Brandon J Aubrey
- Massachusetts General Hospital, Boston, MA 02114, United States; Dana-Farber Cancer Institute, Boston, MA 02115, United States
| | - Traci Blonquist
- Dana-Farber Cancer Institute, Boston, MA 02115, United States
| | | | - Gabriela Hobbs
- Massachusetts General Hospital, Boston, MA 02114, United States
| | - Steven McAfee
- Massachusetts General Hospital, Boston, MA 02114, United States
| | | | - Philip Amrein
- Massachusetts General Hospital, Boston, MA 02114, United States
| | | | - Aura Ramos
- Massachusetts General Hospital, Boston, MA 02114, United States
| | - Emma Logan
- Beth Israel Deaconess Medical Center, Boston, MA 02115, United States
| | - Bimalangshu Dey
- Massachusetts General Hospital, Boston, MA 02114, United States
| | - Thomas Spitzer
- Massachusetts General Hospital, Boston, MA 02114, United States
| | - David Avigan
- Beth Israel Deaconess Medical Center, Boston, MA 02115, United States
| | - Yi-Bin Chen
- Massachusetts General Hospital, Boston, MA 02114, United States
| | - Karen Ballen
- University of Virginia, Charlottesville, VA 22904, United States
| | | | - Amir T Fathi
- Massachusetts General Hospital, Boston, MA 02114, United States
| | - Andrew M Brunner
- Massachusetts General Hospital, Boston, MA 02114, United States.
| |
Collapse
|
48
|
Widjaja Lomanto MY, Wanandi SI, Jayusman AM, Lukmanto D, Prayitno YH, Sutandyo N. Smoking induces different expression of miR-320b and miR-10b-5p in plasma extracellular vesicles of non-small cell lung cancer patients. THE JOURNAL OF LIQUID BIOPSY 2025; 8:100291. [PMID: 40224902 PMCID: PMC11984573 DOI: 10.1016/j.jlb.2025.100291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 04/15/2025]
Abstract
Background Previous studies found that cigarette smoke (CS) exposure could induce NSCLC malignancy and miRNA dysregulation. Yet, the association of CS-induced miRNA dysregulation and NSCLC malignancy has not been clearly understood. This study aimed to evaluate the effect of CS exposure in smokers on the expression of miR-10b-5p and miR-320b in extracellular vesicles (EVs) from NSCLC patients. Material and methods Bioinformatic analysis was conducted to validate miRNA candidates. Blood and tissue samples were collected from NSCLC patients (n = 21) with smoking and non-smoking history. EVs were isolated from plasma and miRNAs were extracted from the isolated EVs. The miRNAs relative expression was analyzed and compared. Results In silico analysis identified miR-320b and miR-10b-5p as potential biomarkers for diagnosing NSCLC in smokers. Experimental analysis revealed differential expression of EVs-associated miRNAs in NSCLC patients with smoking and non-smoking histories. EVs-associated miR-10b-5p was significantly overexpressed in smoker NSCLC patients (p = 0.000), while miR-320b expression was significantly lower in this group (p = 0.018). Additionally, smoking intensity influenced miRNA expression, with higher smoking intensity correlating with increased miR-10b-5p expression and decreased miR-320b expression. ROC analysis demonstrated that EVs were a superior source of miRNAs compared to plasma for NSCLC diagnostics. miR-10b-5p and miR-320b in EVs showed higher diagnostic performance (AUC 0.878; 0.739) compared to plasma (AUC 0.628; 0.559). Conclusion CS exposure induces different expression of miR-10b-5p and miR-320b in EVs of NSCLC patients with smoking history. EV-related miR-10b-5p and miR-320b showed potential to be utilized as prognostic biomarker for smokers NSCLC patients.
Collapse
Affiliation(s)
| | - Septelia Inawati Wanandi
- Master's Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Molecular Biology and Proteomics Core Facilities, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | | | - Donny Lukmanto
- Laboratory of Advanced Vision Sciences, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yuniar Harris Prayitno
- Department of Hematology and Medical Oncology, Dharmais Hospital National Cancer Center, Jakarta, Indonesia
| | - Noorwati Sutandyo
- Master's Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Department of Hematology and Medical Oncology, Dharmais Hospital National Cancer Center, Jakarta, Indonesia
| |
Collapse
|
49
|
Chen B, Yuan S, Wang H. Identification of a cuproptosis‑related prognostic biomarker and therapeutic target in ovarian cancer. Oncol Lett 2025; 29:302. [PMID: 40291472 PMCID: PMC12023028 DOI: 10.3892/ol.2025.15048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/25/2025] [Indexed: 04/30/2025] Open
Abstract
Ovarian cancer (OV) constitutes a significant hazard to the health of women and has low survival and high recurrence rates. Cuproptosis is a newly reported form of copper-dependent regulatory cell death. The present study identified cuproptosis-related long non-coding (lnc)RNAs in OV, highlighting their potential application as prognostic biomarkers and therapeutic targets. The RNA-sequencing data and clinical records of patients with OV were sourced from The Cancer Genome Atlas. Cuproptosis-related lncRNAs were filtered for their prognostic value using univariate and multivariate Cox regression, and least absolute shrinkage selection operator regression. Then, a risk model was formulated using these cuproptosis-related lncRNAs based on correlation coefficients. The risk model was calculated using the following formula: Risk = (0.687927022 × RP11-552D4.1) - (0.659783022 × AP001372.2) - (0.652465319 × RP11-505K9.1) - (1.627006889 × LINC00996). The predictive potential and clinical values of this risk model were identified through survival status, Kaplan-Meier survival curves, immune function, receiver operating characteristic curves, calibration curves, C-index and principal component analysis. Subsequently, the effects of LINC00996 (the lncRNA with the highest correlation coefficient in the risk model) on proliferation, metastasis and sensitivity to cuproptosis were assessed in OV cells. Finally, intracellular location of LINC00996 and the relative regulatory mechanism were predicted. In conclusion, the present study constructed a prognostic risk model based on lncRNAs associated with cuproptosis in OV, which can stratify risk and predict prognosis, and explored the regulatory mechanism of LINC00996 in cuproptosis.
Collapse
Affiliation(s)
- Bingxin Chen
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Shuo Yuan
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Hui Wang
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
50
|
Kannancheri Puthooru D, Yassine M, Gordienko D, Ziental-Gelus N, Desruelles E, Farfariello V, Lemonnier L, Prevarskaya N. Reduced store operated calcium entry contributes to autophagy mediated escape of prostate cancer to oxaliplatin treatment. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119953. [PMID: 40189057 DOI: 10.1016/j.bbamcr.2025.119953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 03/21/2025] [Accepted: 03/28/2025] [Indexed: 04/17/2025]
Abstract
Oxaliplatin, a third-generation platinum-based chemotherapeutic drug, induces cell cycle arrest and apoptosis in prostate cancer treatment. However, both intrinsic and acquired resistance mechanisms limit its therapeutic efficacy. Notably, chemotherapeutic agents often induce autophagy-a cellular recycling process-that can contribute to drug resistance. Calcium (Ca2+) signalling plays a pivotal role in regulating cell fate. However, the involvement of Ca2+ and Ca2+ channels in oxaliplatin resistance within prostate cancer cells remains controversial and poorly understood. In this study, we demonstrate that oxaliplatin treatment enhances autophagy in prostate cancer cells. Concurrently, oxaliplatin modulates the expression of key proteins involved in store-operated calcium entry (SOCE): it upregulates Orai3 channels while downregulating Orai1 and Stim1. These alterations result in diminished SOCE activity, contributing to an apoptosis-resistant phenotype. Importantly, we found that targeting Orai3 expression and inhibiting autophagy sensitizes prostate cancer cells to oxaliplatin-induced apoptosis. Our findings suggest that combining Orai3 downregulation with autophagy inhibition may enhance the efficacy of oxaliplatin in treating prostate cancer. This combinatorial approach could hold potential for overcoming resistance and improving therapeutic outcomes.
Collapse
Affiliation(s)
- Dheeraj Kannancheri Puthooru
- Université de Lille, Inserm, U1003 - PHYCEL - Physiologie Cellulaire, LabEx "Ion Channel Science and Therapeutics", F-59000, Lille, France
| | - Maya Yassine
- Université de Lille, Inserm, U1003 - PHYCEL - Physiologie Cellulaire, LabEx "Ion Channel Science and Therapeutics", F-59000, Lille, France
| | - Dmitri Gordienko
- Université de Lille, Inserm, U1003 - PHYCEL - Physiologie Cellulaire, LabEx "Ion Channel Science and Therapeutics", F-59000, Lille, France
| | - Nathalie Ziental-Gelus
- Université de Lille, Inserm, U1003 - PHYCEL - Physiologie Cellulaire, LabEx "Ion Channel Science and Therapeutics", F-59000, Lille, France
| | - Emilie Desruelles
- Université de Lille, Inserm, U1003 - PHYCEL - Physiologie Cellulaire, LabEx "Ion Channel Science and Therapeutics", F-59000, Lille, France
| | - Valerio Farfariello
- Université de Lille, Inserm, U1003 - PHYCEL - Physiologie Cellulaire, LabEx "Ion Channel Science and Therapeutics", F-59000, Lille, France
| | - Loïc Lemonnier
- Université de Lille, Inserm, U1003 - PHYCEL - Physiologie Cellulaire, LabEx "Ion Channel Science and Therapeutics", F-59000, Lille, France.
| | - Natalia Prevarskaya
- Université de Lille, Inserm, U1003 - PHYCEL - Physiologie Cellulaire, LabEx "Ion Channel Science and Therapeutics", F-59000, Lille, France.
| |
Collapse
|