51
|
Qiao R, Zhou X, Li W, Zhong R, Wang J, Song Y, Zhang J, Xu T, Wang Y, Dai L, Gu W, Han B, Yang R. The association between blood-based HYAL2 methylation and early-stage lung cancer: a case-control study. Lung Cancer Manag 2025; 14:2477411. [PMID: 40168159 PMCID: PMC11970770 DOI: 10.1080/17581966.2025.2477411] [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: 07/29/2024] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Blood-based DNA methylation biomarkers have great potential for the early detection of lung cancer (LC). Here, we investigated the association between HYAL2 methylation in peripheral blood and LC. METHODS Matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) mass spectrometry was performed to measure the methylation levels of 4 CpG sites in HYAL2 gene in two independent case-control studies (168 LC cases and 167 controls in Study I, 677 LC cases and 833 controls in Study II). Logistic regression adjusted for covariates was conducted for odds ratios (ORs) and 95% confidence intervals (CIs). Non-parametric tests were applied for the comparisons of stratified groups. RESULTS Hypomethylation of all 4 CpG sites in HYAL2 was associated with early-stage LC in the two studies (ORs range from 1.91 to 3.07 in Study I, ORs range from 1.39 to 1.86 in Study II, p < 0.05 for all). The associations were still significant for the very early-stage LC patients (stage I). Subgroup analysis indicated that the associations could be enhanced by male gender and older age. Moreover, decreased HYAL2 methylation was correlated with increased tumor size, tumor length and stage. CONCLUSIONS Our results suggested blood-based HYAL2 hypomethylation as a potential biomarker for LC early detection.
Collapse
Affiliation(s)
- Rong Qiao
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Xiajie Zhou
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
- Nanjing Medical University Affiliated Nanjing Center for Disease Control and Prevention, Nanjing, China
| | - Wenli Li
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Runbo Zhong
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jun Wang
- Research and Development, Nanjing TANTICA Biotechnology Co. Ltd, Nanjing, China
| | - Yakang Song
- Research and Development, Nanjing TANTICA Biotechnology Co. Ltd, Nanjing, China
| | - Jing Zhang
- Research and Development, Nanjing TANTICA Biotechnology Co. Ltd, Nanjing, China
| | - Tian Xu
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yue Wang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Wanjian Gu
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Baohui Han
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Rongxi Yang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
- Research and Development, Nanjing TANTICA Biotechnology Co. Ltd, Nanjing, China
| |
Collapse
|
52
|
Li S, Liu T, Li C, Zhang Z, Zhang J, Sun D. Overcoming immunotherapy resistance in colorectal cancer through nano-selenium probiotic complexes and IL-32 modulation. Biomaterials 2025; 320:123233. [PMID: 40081224 DOI: 10.1016/j.biomaterials.2025.123233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/11/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND AND OBJECTIVE Colorectal cancer (CRC) is a major global health burden, with immunotherapy often limited by immune tolerance and resistance. This study introduces an innovative approach using Selenium Nanoparticles-Loaded Extracellular Vesicles combined with Interleukin-32 and Engineered Probiotic Escherichia coli Nissle 1917 (SeNVs@NE-IL32-EcN) to enhance CD8+ T cell-mediated immune responses and overcome immunotherapy resistance. METHODS Single-cell RNA sequencing (scRNA-seq) and transcriptomic analyses were performed to identify key immune cells and regulators involved in CRC immunotherapy resistance, focusing on CD8+ T cells and the regulatory factor IL32. A humanized xenograft mouse model was used to evaluate the impact of IL32 and SeNVs@NE-IL32-EcN on tumor growth and immune responses. The SeNVs@NE-IL32-EcN complex was synthesized through a reverse micelle method and functionalized using extracellular vesicles. Its morphology, size, antioxidant activity, and safety were characterized using electron microscopy, dynamic light scattering (DLS), and in vitro co-culture assays. RESULTS Single-cell analyses revealed a significant reduction in CD8+ T cell infiltration in immunotherapy-resistant CRC patients. IL32 was identified as a key regulator enhancing CD8+ T cell cytotoxic activity through granzyme B and IFN-γ secretion. Treatment with SeNVs@NE-IL32-EcN significantly improved the proliferation and activity of CD8+ T cells and reduced tumor progression in humanized mouse models. In vitro and in vivo results demonstrated the complex's biocompatibility, antioxidant properties, and ability to enhance CRC immunotherapy while mitigating immune tolerance. CONCLUSION SeNVs@NE-IL32-EcN offers a novel nano-biomaterial strategy that integrates nanotechnology and probiotic therapy to enhance CD8+ T cell-mediated immunity and overcome CRC immunotherapy resistance. This study lays the foundation for future therapeutic applications in cancer treatment by advancing immune-modulating biomaterials.
Collapse
Affiliation(s)
- Shiquan Li
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Tao Liu
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Chenyao Li
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Zhiyuan Zhang
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Jiantao Zhang
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Di Sun
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
53
|
Bajgai J, Jun M, Oh JH, Lee JH. A perspective on the potential use of aptamer-based field-effect transistor sensors as biosensors for ovarian cancer biomarkers CA125 and HE4. Talanta 2025; 292:127954. [PMID: 40120511 DOI: 10.1016/j.talanta.2025.127954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 02/25/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Ovarian cancer (OC) is one of the most fatal gynaecological malignancies, primarily because of its typically asymptomatic early stages, which complicates early detection. Therefore, developing sensitive and appropriate biomarkers for efficient diagnosis of OC is urgently needed. Aptamers, short sequences of single-stranded DNA or RNA molecules, have become crucial in tumor diagnosis because of their high affinity for specific molecules produced by tumors. This ability allows aptamers to accurately detect OC, thus providing better survival rates and a reduced disease burden. Biosensors that combine recognition molecules and nanomaterials are essential in various fields, including disease diagnosis and health management. Molecular-specific field-effect transistor (FET) biosensors are particularly promising due to their rapid response times, ease of miniaturization, and high sensitivity in detecting OC. Aptamers, which are known for their stability and structural tunability, are increasingly being used as biological recognition units in FET biosensors, offering selective and high-affinity binding to target molecules that are ideal for medical diagnostics. This review explores the recent advancements in biosensors for OC detection, including FET biosensors with aptamer-functionalized nanomaterials for CA125 and HE4. Furthermore, this review provides an overview of the structure and sensing principles of these advanced biosensors, preparation methods and functionalization strategies that enhance their performance. Additionally, notable progress and potential of biosensors, including aptamer-functionalized FET biosensors for OC diagnosis have been summarized, emphasising their role and clinical validation in advancing medical diagnostics and improving patient outcomes through enhanced detection capabilities.
Collapse
Affiliation(s)
- Johny Bajgai
- Department of Laboratory Medicine, Yonsei University Wonju College of Medicine Wonju, Gangwon-do 26426, Republic of Korea; Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Minsang Jun
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Republic of Korea
| | - Joon Hak Oh
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Republic of Korea
| | - Jong-Han Lee
- Department of Laboratory Medicine, Yonsei University Wonju College of Medicine Wonju, Gangwon-do 26426, Republic of Korea; Research Institute of Metabolism and Inflammation, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea.
| |
Collapse
|
54
|
Li L, Lan Z, Qiao H, Meng X, Shi Z, Zhang W, Wang Y, Sun Z, Cui Q, Wang L, Zhou S, Hu F, Zhang D, Dai Y, Chen H, Geng Y. Design of NanoBiT-Nanobody-based FGL1 biosensors for early assisted diagnosis of esophageal cancer. Biomaterials 2025; 320:123286. [PMID: 40138964 DOI: 10.1016/j.biomaterials.2025.123286] [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: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Esophageal cancer (EC) is one of the most common causes of cancer-related mortality due in part to challenges in early diagnosis. Biomarker identification is crucial for improved early screening and treatment strategies for patients. Firstly, we employed serum proteomics techniques to screen for potential biomarkers in 15 early-stage EC patients and 5 healthy individuals. Among the differentially expressed proteins, FGL1 emerged as a promising candidate (AUC = 0.974) for early detection of EC. Subsequently, we developed NanoBiT-conjugated dual nanobodies (NBNB) sensors for robust and quantitative signal detection in fetal bovine serum (FBS) in 30 min or less, with a limit of detection (LoD) of 11.38 pM. In a case-control study recruiting 96 EC patients and 99 control samples, testing serum samples with the developed NBNB sensors revealed significantly elevated serum level of FGL1 in all-stage EC patients (AUC = 0.7880) and early-stage EC patients (AUC = 0.8286). Additionally, the combined diagnostic performance of FGL1 and CEA in EC samples is notably enhanced (AUC = 0.8847). These findings propose FGL1 as a novel and promising target for the early-stage EC diagnosis and treatment selection. Furthermore, we applied the assay to patients across six types of cancer, suggesting FGL1 as a potential pan-cancer marker. This study introduces a rapid, easy-to-use, cost-effective, reliable, universal, and high-throughput alternative to meet the growing demand for cancer biomarker testing in both academic and clinical settings.
Collapse
Affiliation(s)
- Lingyun Li
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhongyun Lan
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Huarui Qiao
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiangjing Meng
- Shandong Provincial Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China
| | - Ziyang Shi
- Queen Mary University of London Engineering School, Northwestern Polytechnical University, Xi'an, 710129, China
| | - Wanting Zhang
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yi'ang Wang
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zengchao Sun
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qianqian Cui
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lu Wang
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Siyu Zhou
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fangzheng Hu
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Daizhou Zhang
- Shandong Provincial Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan, 250101, China.
| | - Yuanyuan Dai
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital of Chinese Academy of Medical Sciences Langfang Campus, Langfang, 065001, China.
| | - Hao Chen
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Yong Geng
- State Key Laboratory of Drug Research, The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
55
|
Neto LPM, Dos Santos L, Carvalho LFCS, Santos ABO, Martin AA, Canevari RA. Integrating Raman spectroscopy and RT-qPCR for enhanced diagnosis of thyroid lesions: A comparative study of biochemical and molecular markers. J Pharm Biomed Anal 2025; 261:116844. [PMID: 40179617 DOI: 10.1016/j.jpba.2025.116844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
Thyroid cancer is the most prevalent endocrine malignancy, with increasing incidence due to advancements in diagnostic techniques. Ultrasound (US) and fine needle aspiration (FNA) cytology, widely used in clinical practice, have detection accuracies ranging from 65 % to 95 %. However, these methods may yield inconclusive or difficult-to-interpret results, emphasizing the need for complementary diagnostic techniques. This study explores the integration of Raman spectroscopy and gene expression analysis via RT-qPCR to improve the diagnosis of thyroid lesions, classified into groups: follicular thyroid carcinoma (FTC), papillary thyroid carcinoma (PTC) and goiter tissues. Healthy tissue samples were used as normalizing controls in both analysis. Raman spectroscopy analyzed 35 samples, while RT-qPCR assessed 33 samples. For comparison, the same 19 samples previously analyzed by both techniques were examined. Raman spectroscopy, a non-invasive technique, has shown effectiveness in distinguishing between benign and malignant thyroid tissues by identifying key biochemical components such as DNA, RNA, proteins, and lipids. The distinguishing of FTC from goiter using Raman spectroscopy achieved an accuracy rate of 82.3 %. Gene expression analysis via RT-qPCR focused on six genes: TG, TPO, PDGFB, SERPINA1, TFF3, and LGALS3. Specifically, SERPINA1 was overexpressed in PTC, TFF3 showed elevated levels in FTC, and LGALS3 was elevated in both PTC and FTC compared to goiter and normal tissues. These findings align with existing literature, suggesting that these genes could serve as valuable diagnostic molecular markers. The expression analysis of these genes within this subset of samples demonstrated concordance with the classification derived from PCA of Raman spectroscopy data. The integration of Raman spectroscopy and RT-qPCR offers a complementary approach to traditional histological analysis, providing enhanced sensitivity and specificity in diagnosing thyroid lesions.
Collapse
Affiliation(s)
- Lázaro P M Neto
- Universidade do Vale do Paraíba, UNIVAP, Instituto de Pesquisa e Desenvolvimento, Avenida Shishima Hifumi 2911, Urbanova, São José dos Campos, São Paulo 12244-000, Brazil; Pontifícia Universidade Católica, PUC Minas, Departamento de Ciências Biológicas e da Saúde, Avenida Padre Cletus Francis Cox, 1661, Jardim Country Club, Poços de Caldas, Minas Gerais 37714-620, Brazil
| | - Laurita Dos Santos
- Universidade do Vale do Paraíba, UNIVAP, Instituto de Pesquisa e Desenvolvimento, Avenida Shishima Hifumi 2911, Urbanova, São José dos Campos, São Paulo 12244-000, Brazil; Universidade Brasil, Instituto de Pesquisa, Campus São Paulo, Rua Carolina Fonseca, 235, Vila Santana, SP, São Paulo 08230-030, Brazil
| | | | - André B O Santos
- Universidade de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Avenida Doutor Arnaldo, 455, Cerqueira César, SP, São Paulo 01246-903, Brazil
| | - Airton A Martin
- Universidade do Vale do Paraíba, UNIVAP, Instituto de Pesquisa e Desenvolvimento, Avenida Shishima Hifumi 2911, Urbanova, São José dos Campos, São Paulo 12244-000, Brazil; Universidade Brasil, Instituto de Pesquisa, Campus São Paulo, Rua Carolina Fonseca, 235, Vila Santana, SP, São Paulo 08230-030, Brazil
| | - Renata A Canevari
- Universidade do Vale do Paraíba, UNIVAP, Instituto de Pesquisa e Desenvolvimento, Avenida Shishima Hifumi 2911, Urbanova, São José dos Campos, São Paulo 12244-000, Brazil.
| |
Collapse
|
56
|
Gupta AK, Sahu Y, Pal D, Kumar N, Jain SK. Assessing novel analogues of nilutamide as a human androgen receptor antagonist: A detailed investigation of drug design using a bioisosteric methodology including ADMET profiling, molecular docking studies and molecular dynamics simulation. Comput Biol Chem 2025; 117:108424. [PMID: 40112513 DOI: 10.1016/j.compbiolchem.2025.108424] [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: 12/31/2024] [Revised: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Cancer is a significant health and economic concern worldwide. Prostate cancer (PC) ranks as the fourth leading cause of global death and is the second most prevalent malignancy in males. Androgens are essential for the progress and growth of the prostate gland. PC is caused by androgens binding to receptors, which activates genes that promotes the development of PC. Nilutamide (NLM) is an antiandrogen medicine used in the treatment of PC. However, throughout treatment, it induces various toxicities and leads to resistance in patients. The objective of the work was to designed and evaluated safer NLM analogues using computational approaches with optimized pharmacokinetic profiles and less toxicity. Newer bioisosteres of the designed NLM analogues and their ADMET scores were calculated using the MolOpt and ADMETlab 3.0 tools, respectively. We conducted docking investigations of the designed ligands using AutoDock Vina software. The MolOpt web server produces 1575 bioisosteres of NLM using the scaffold transformation method. The 47 bioisosteres were selected based on pharmacokinetic profiles, drug likeness (DL) and drug score (DS) prediction scores and were determined to be optimum to excellent in comparison to NLM. The analogues NLM28, NLM31, NLM34, NLM38, NLM40, NLM44, NLM45, and NLM47 exhibited favorable interactions and docking scores with the protein (PDB ID: 2AM9). The molecular dynamics (MD) simulation results revealed that the NLM34 and NLM40 complexes were found stable during the 100 ns run. The findings indicate that the NLM analogues, particularly NLM34 and NLM40 have the potential to be used as promising antiandrogen agents for PC therapy.
Collapse
Affiliation(s)
- Ajay Kumar Gupta
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh 495009, India
| | - Yogita Sahu
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh 495009, India
| | - Dipti Pal
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh 495009, India
| | - Neeraj Kumar
- Department of Pharmaceutical Chemistry, Bhupal Nobles' College of Pharmacy, Udaipur, Rajasthan 313001, India
| | - Sanmati Kumar Jain
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh 495009, India.
| |
Collapse
|
57
|
Lei X, Wang F, Zhang X, Huang J, Huang Y. The potential mechanisms by which Xiaoyao Powder may exert therapeutic effects on thyroid cancer were examined at various levels. Comput Biol Chem 2025; 117:108412. [PMID: 40056710 DOI: 10.1016/j.compbiolchem.2025.108412] [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: 12/06/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Thyroid cancer (TC) is the most prevalent endocrine malignancy, with a rising incidence necessitating safer treatment strategies to reduce overtreatment and its side effects. Xiaoyao Powder (XYP), a widely used herbal formula, shows promise in treating TC. This study aims to investigate the mechanisms by which XYP may affect TC. METHODS The components of XYP were identified through database retrieval, and targets related to TC were collected to construct a target network for key screening. GEO dataset samples analyzed immune cells and identified significantly differentially expressed core genes (SDECGs). Based on SDECG expression and clustering, samples were classified for comparison. WGCNA was employed to identify gene modules linked to clinical characteristics. ML models screened characteristic genes and constructed a nomogram validated using another GEO dataset. MR methods explored causal relationships between genes and TC. RESULTS The top ten active components of XYP were identified, along with 27 SDECGs that exhibited significant differences in immune cell infiltration between TC patients and normal controls. The nomogram effectively predicted TC risk, validated through ROC curves. Key characteristic genes included SMIM1, PPP1R16A, KIAA1462, DNAJC22, and EFNA5. CONCLUSION XYP may treat TC by regulating SMIM1, PPP1R16A, KIAA1462, DNAJC22, EFNA5, and associated immune pathways; this provides theoretical support for its potential mechanisms.
Collapse
Affiliation(s)
- Xiaoli Lei
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Feifei Wang
- Department of Quality Control, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, China
| | - Xinying Zhang
- Department of Endocrinology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiaxi Huang
- Department of Pharmacy, Huoqiu County First People's Hospital, Liuan, China
| | - Yanqin Huang
- Department of Endocrinology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
58
|
Nugteren S, Wang H, van Kooten C, Gelderman KA, Trouw LA. Autoantibodies and therapeutic antibodies against complement factor H. Immunol Lett 2025; 274:107002. [PMID: 40118156 DOI: 10.1016/j.imlet.2025.107002] [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: 12/24/2024] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
The complement system is a crucial part of our immune defense as, upon recognition, it can kill pathogens fast and effectively. However, misguided complement activation could cause damage to host tissues. Therefore, a well-controlled regulation of the complement system is a necessity to prevent collateral damage. Regulation is achieved by several complement inhibitory proteins, acting at different levels of the complement system. One of these complement regulators is factor H, the main regulator of the alternative complement activation pathway. Factor H can regulate the complement system both in fluid-phase and on the host cell surface by, for example, acting as co-factor for factor I, inactivating C3b. The functional properties of factor H are located within different regions of the protein. Functional impairment of factor H, either because of genetic variants, competing proteins such as the factor H-related proteins and proteins from certain pathogens, but also the presence of autoantibodies will impact on complement activation. However, exact consequences are dependent on the region within factor H that is affected. Autoantibodies binding to factor H have been shown to inhibit several regulatory functions of factor H, which is observed in diseases such as membranoproliferative glomerulonephritis and atypical hemolytic uremic syndrome. As more recently the presence of anti-factor H autoantibodies has also been discovered in several other diseases, ranging from autoimmune diseases to cancer, this review provides an overview of the presence of factor H autoantibodies described in these diseases. Factor H autoantibodies are reported to have inhibitory, or enhancing, effects on factor H, depending on the epitopes that are recognized. Formal conclusions about the pathogenicity of the factor H autoantibodies in some of these diseases cannot be drawn yet. Importantly, understanding the binding and functional impact of anti-factor H (auto)antibodies will allow targeted interventions to diminish pathological consequences of anti-factor H autoantibodies but may also open up additional avenues for the use of anti-factor H antibodies as therapeutic agents.
Collapse
Affiliation(s)
- Saskia Nugteren
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Haiyu Wang
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Cees van Kooten
- Department of Internal Medicine section Nephrology, Center of Expertise for Lupus, Vasculitis and Complement- mediated Systemic Autoimmune Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Kyra A Gelderman
- Erasmus Medical Center, Department of Immunology, Laboratory Medical Immunology, Rotterdam, the Netherlands
| | - Leendert A Trouw
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
59
|
van der Merwe M, Myburgh K, Garnis C, Towle R, Engelbrecht AM. Unravelling the role of extracellular vesicles in cervical cancer: Mechanisms of progression, resistance, and emerging therapeutic strategies. Gene 2025; 957:149467. [PMID: 40204037 DOI: 10.1016/j.gene.2025.149467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/18/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Cervical cancer remains a significant global health challenge, particularly in its advanced stages, where treatment resistance complicates effective management. Extracellular vesicles (EVs) are crucial mediators of tumor progression and resistance, primarily through the transfer of miRNA cargo. In cervical cancer, specific miRNAs, including oncogenic miRNAs such as miR-21, miR-221-3p, miR-486-5p, and miR-92a-3p are upregulated in both cells and EVs, promoting proliferation, migration, epithelial-to-mesenchymal transition (EMT), and immune evasion-all of which contribute to therapy resistance and an aggressive tumor phenotype. Conversely, tumor-suppressive miRNAs, such as miR-122-5p, miR-100, and miR-142-3p, are selectively exported from cancer cells via EVs, suggesting a protective mechanism by which cancer cells eliminate these tumor suppressors. This review focuses on the role of oncogenic and tumor-suppressive miRNAs within EVs and their implications for cervical cancer progression and treatment resistance. Additionally, it examines the dynamic interactions between the tumor microenvironment (TME) and EV cargo, as well as emerging EV-based therapeutic strategies. These include the encapsulation of chemotherapeutic agents within EVs, the use of anti-miRs to silence oncogenic miRNAs, the delivery of tumor-suppressive miRNAs, the inhibition of EV release, and the targeting of downstream miRNA-regulated proteins. While miRNA-based therapies remain in the early stages, they hold significant promise for overcoming treatment resistance and improving cervical cancer outcomes.
Collapse
Affiliation(s)
- Michelle van der Merwe
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa.
| | - Kathy Myburgh
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Cathie Garnis
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z1L3, Canada
| | - Rebecca Towle
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, BC V5Z1L3, Canada
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
60
|
Zheng W, Liu Y, Liu J, Zhao Y, Wang P, Wang Y, Wang Y, Lu Z, Liu X, Shi Y, Zhou N, González FE, Sun H, Zhou D, Xu X. Copper/calcium co-doped carbon dots for targeted cancer therapy with dual-mode imaging and synergistic induction of cuproptosis and calcium-mediated apoptosis. J Colloid Interface Sci 2025; 690:137337. [PMID: 40117884 DOI: 10.1016/j.jcis.2025.137337] [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: 01/13/2025] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Squamous cell carcinoma remains a highly aggressive malignancy with persistently high global incidence and mortality rates, posing significant challenges for effective treatment. Traditional chemotherapies lack specificity, leading to damage in normal tissues and severe side effects, highlighting the urgent need for targeted therapeutic strategies. In this study, copper and calcium co-doped carbon dots (Cu/Ca-CDs) were synthesized using a vacuum-confined heating method. These Cu/Ca-CDs demonstrated excellent tumor-targeting ability through specific binding to folate receptors on murine squamous cell carcinoma cell line (SCC7), facilitated by their pterin ring structure. Mechanistic studies revealed that Cu/Ca-CDs induced SCC7 tumor cell death through copper-induced cuproptosis and calcium overload-mediated apoptosis, as confirmed by Western blot, immunofluorescence staining, and Rhod-2 calcium probe analyses. The dual-mode imaging capability of Cu/Ca-CDs, enabled by fluorescence and computed tomography properties, allowed for real-time tracking of their distribution and accumulation within tumors. This imaging-guided approach ensured precise delivery to tumor tissues while minimizing damage to normal tissues. In vivo experiments demonstrated significant tumor volume reduction and increased survival rates in tumor-bearing mice treated with Cu/Ca-CDs, without any observed toxicity to normal tissues or changes in body weight, underscoring the efficacy and biosafety of Cu/Ca-CDs. These findings highlight Cu/Ca-CDs as a promising strategy for precision oncology, offering effective tumor targeting, dual-mode imaging, and synergistic anti-tumor efficacy with reduced side effects.
Collapse
Affiliation(s)
- Wenqian Zheng
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Yang Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Jinru Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Yuping Zhao
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Peiyu Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Yi Wang
- Graduate Program in Applied Physics, Northwestern University, Evanston, IL 60208, USA
| | - Yuxuan Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Zeyu Lu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Xiaofan Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Yaru Shi
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Na Zhou
- School of Pharmacy, State Key Laboratory of Quality Research in Chinese Medicines and Laboratory of Drug Discovery from Natural Resources and Industrialization, Macau University of Science and Technology, Macau 999078, PR China
| | - Fermín Eduardo González
- Laboratory of Experimental Immunology & Cancer, Faculty of Dentistry, University of Chile, Santiago 8380492, Chile
| | - Hongchen Sun
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Ding Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, PR China.
| | - Xiaowei Xu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, PR China.
| |
Collapse
|
61
|
Huang Y, Zeng Y, Cui S, Liu C, Cai X. Geometric a priori informed bent-ray tracing for accelerated sound speed imaging in ultrasound computed tomography. ULTRASONICS 2025; 151:107595. [PMID: 40081289 DOI: 10.1016/j.ultras.2025.107595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/25/2025] [Accepted: 02/04/2025] [Indexed: 03/15/2025]
Abstract
Bent-ray tracing ultrasound computed tomography (USCT) is a promising technique for breast cancer screening which quantitatively provides speed-of-sound (SOS) distribution in human breasts. In this modality, SOS images are reconstructed with an iterative process to match the measured time-of-flights and the ones predicted by Eikonal equation solved with the fast marching method (FMM). The Eikonal equation is meant to be applied in SOS heterogeneous media and its evaluation with FMM is an computational expensive process. However, in USCT, the object is placed in a homogeneous coupling medium. Thus, the acoustic environment is formed by two parts, the homogeneous background (coupling medium) and the heterogeneous object. In this work, we leverage this strong a priori information and propose a method to accelerate SOS image formation for bent-ray tracing USCT. We show that, given the boundary information of the object, Eikonal equation only needs to be evaluated in a limited area covering the object. For that, the partial FMM and the associated ray-tracing strategy are proposed to reduce the computational cost of the forward modeling. We also managed to restrict image reconstruction area inside the object for improved convergence rate of the optimization. Both the simulation and phantom imaging experiments with ring transducer arrays demonstrated that the proposed method reduces the reconstruction time in an object size dependent manner. For the object occupying 20.3% to 56.3% of the image field of the ring array, we observed 30.1%-61.9% reduction in image reconstruction time without sacrificing the image quality, compared to classical method. The proposed strategy can be adopted for fast SOS imaging with bent-ray tracing USCT to improve patient throughput for breast cancer screening.
Collapse
Affiliation(s)
- Yiming Huang
- School of Information Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yi Zeng
- School of Information Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Shilong Cui
- School of Information Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Chengcheng Liu
- Academy for Engineering and Technology, Fudan University, Shanghai, 200433, China.
| | - Xiran Cai
- School of Information Science and Technology, ShanghaiTech University, Shanghai, 201210, China; Shanghai Engineering Research Center of Intelligent Vision and Imaging, ShanghaiTech University, Shanghai, 201210, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
62
|
Wang Y, Jiang N, Wu S, Lin G, Lu W, Shang B, Zhu X, Han W, Li J, Chen Y. An injectable hydrogel for hemostasis and tumor suppression in intraoperative breast cancer. BIOMATERIALS ADVANCES 2025; 172:214219. [PMID: 39987716 DOI: 10.1016/j.bioadv.2025.214219] [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: 07/22/2024] [Revised: 01/14/2025] [Accepted: 02/02/2025] [Indexed: 02/25/2025]
Abstract
In the period between surgery and systemic therapy for breast cancer, residual tumor cells may proliferate, leading to tumor recurrence. Additionally, intraoperative wound bleeding may cause surgical failure or the spread of tumor cells. This study introduces an innovative injectable hydrogel composed of oxidized hyaluronic acid (OHA) loaded 5-fluorouracil (5-FU) and N-carboxyethyl chitosan (CEC), designed for intraoperative hemostasis and tumor suppression in intraoperative breast cancer. The CEC/OHA injectable hydrogel was synthesized through a Schiff base reaction between the aldehyde group of OHA and the amino group of CEC, incorporating 5-FU during hydrogel formation. This CEC/OHA injectable hydrogel demonstrated hemostatic effects comparable to gelatin sponges in both an in vivo rat liver hemorrhage model and an in vitro rat tail amputation model. When loaded with 5-FU, the injectable hydrogel effectively inhibited the proliferation of MDA-MB-231 breast cancer cells in vitro, significantly inhibited tumor growth and recurrence in vivo, and did not induce significant damage or inflammatory response in any major organ. This CEC/OHA & 5-FU injectable hydrogel is envisioned as a complementary therapeutic regimen during the intraoperative period in breast cancer surgery to prevent hemostasis and tumor recurrence.
Collapse
Affiliation(s)
- Yue Wang
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Nan Jiang
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Shuhan Wu
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Guangshuai Lin
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Wudang Lu
- Xi'an Libang Pharmaceutical Co., Ltd, Xi'an, Shaanxi 710075, China
| | - Bin Shang
- Xi'an Libang Pharmaceutical Co., Ltd, Xi'an, Shaanxi 710075, China
| | - Xulong Zhu
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Wei Han
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Jianhui Li
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China.
| | - Yongmei Chen
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science & Technology, Xi'an, Shaanxi 710021, China.
| |
Collapse
|
63
|
Bahadoran E, Mohammadi D, Jalilvand M, Moghbelinejad S. Evaluation of CircHIPK3 biomarker potential in breast cancer. Pract Lab Med 2025; 45:e00470. [PMID: 40226123 PMCID: PMC11984561 DOI: 10.1016/j.plabm.2025.e00470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/14/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Background Nowadays, the investigation of circular RNAs (circRNAs) in various cancers is of great interest. In this research, we evaluated circHIPK3 biomarker potential in breast cancer (BC). Methods The studied samples were 100 cancer and adjacent normal tissues, plasma from 95 cancer patients, 42 patients with fibroadenomatosis, and 93 healthy donors. Illumina high-throughput Hi Seq 2000 sequencing performed expression profiling on 4 pairs of cancerous and normal breast tissues. For expression confirmation, Quantitative real-time fluorescent polymerase chain reaction (qRT-PCR) was used to detect the expression level of circHIPK3. CircHIPK3 diagnostic efficacy was evaluated by the receiver operating characteristic curve (ROC). Results Based on high-throughput sequencing and bioinformatics results circHIPK3 had the highest expression in cancer tissues (P = 0.00034). Real-time results showed expression upregulation of circHIPK3 in BC tissues and plasma in comparison to healthy controls (P < 0.0001). For diagnostic potential, the area under the curve (AUC) result was 0.8087 (95 % CI: 0.7309 to 0.8866, P < 0.0001). Also, our results showed high specificity and sensitivity of circHIPK3 when evaluated alongside the CA-15-3 and CEA. Pathologic criteria evaluation showed that upregulation of circHIPK3 correlates with tumor size. Conclusions CircHIPK3 is significantly upregulated in BC tissues and plasma compared to healthy controls, demonstrating high diagnostic potential with an AUC of 0.8087. The expression of circHIPK3 correlates with tumor size, indicating its relevance in the pathologic assessment of BC.
Collapse
Affiliation(s)
- Ensiyeh Bahadoran
- Cellular and Molecular Research Centre, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Davood Mohammadi
- Department of Surgery, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Manijeh Jalilvand
- Cellular and Molecular Research Centre, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sahar Moghbelinejad
- Cellular and Molecular Research Centre, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
64
|
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
|
65
|
Li B, Liu T, Zhao M, Guo D, Peng W, Tao Z, Hu X, Jin J. MYC amplification sensitizes TNBC to CHK1 inhibitors. Cell Signal 2025; 131:111709. [PMID: 40037423 DOI: 10.1016/j.cellsig.2025.111709] [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: 02/09/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, for which effective therapy is urgently needed. We demonstrated that MYC overexpression was associated with TNBC subtype and promoted the cell proliferation, invasion and migration in TNBC cells. Moreover, MYC overexpression induced replication stress and DNA damage in TNBC cells. Our subsequent results revealed that the novel second-generation CHK1 inhibitor, prexasertib, exhibited a more pronounced inhibitory effect in MYC-overexpressed TNBC cells compared to other DNA damage repair inhibitors, including ATR, WEE1, and PARP inhibitors. Prexasertib induced synergistic lethality with MYC-overexpressed TNBC cells by generating excessive DNA damage. Intriguingly, RNA-seq analysis identified an increase in MYC levels and activation of MYC-related pathways following prexasertib treatment, while western blot results showed that prexasertib led to MYC protein degradation independent of proteasome pathway. In addition, MYC overexpression was associated with an immunosuppressive microenvironment and high PD-L1 expression. Prexasertib activated cGAS-STING pathway by inducing DNA damage. Therefore, combination of prexasertib and immune checkpoint inhibitors will be a potential therapeutic strategy for MYC-overexpressed TNBC. In conclusion, our findings demonstrated that MYC overexpression characterizes an aggressive TNBC subtype, enabling synergistic lethality with CHK1 inhibitors. CHK1 inhibitors will be a potential therapeutic strategy in TNBC patients with MYC overexpression.
Collapse
Affiliation(s)
- Bin Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tiantian Liu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mingchuan Zhao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Duancheng Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wenxia Peng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhonghua Tao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Juan Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
66
|
Schnabl SD, Klubien J, O'Rourke CJ, Bull Nordkild S, Kugler JM, Dam Nielsen S, Andersen JB, Pommergaard HC. Validation of Two Prognostic Gene Scores in Patients Undergoing Liver Resection for Hepatocellular Carcinoma. J Clin Exp Hepatol 2025; 15:102544. [PMID: 40248345 PMCID: PMC12002650 DOI: 10.1016/j.jceh.2025.102544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/04/2025] [Indexed: 04/19/2025] Open
Abstract
Background/Aims Several prognostic gene signatures have been proposed as predictors of the prognosis of hepatocellular carcinoma (HCC), yet none are implemented in the clinical setting. We aimed to validate two gene scores previously derived from European cohorts. Methods The patients who underwent liver resection for HCC at Copenhagen University Hospital, Rigshospitalet from 2014 to 2018 were included. RNA sequencing determined the expression of genes in the '5-gene score' (HN1, RAN, RAMP3, KRT19, TAF9B) and 'HepatoPredict' (CLU, DPT, SPRY2, CAPSN1). Univariable Cox regression assessed associations with overall and disease-free survival. These parameters were also analyzed in the The Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC) (n = 359) and National Institute of Health (NIH) (n = 178) cohorts. Results Among 51 patients (88% male), 59% had no underlying liver disease and 25% had cirrhosis. No individual genes were significantly associated with overall survival in the Danish cohort. In the TCGA-LIHC cohort, CLU was linked to better overall survival, and in the NIH cohort, high expression of SPRY2 was associated with poorer overall survival. In the TCGA-LIHC cohort, HN1, RAN, and TAF9B were associated with poorer overall survival, while RAMP3 was linked to better overall survival. No genes were associated with disease-free survival. Conclusion Few individual genes significantly predicted survival in the larger cohorts, and none in the Danish cohort. However, the clinical implication of this needs further investigation.
Collapse
Affiliation(s)
- Stinna D. Schnabl
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Hepatic Malignancy Surgical Research Unit (HEPSURU), Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Jeanett Klubien
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Hepatic Malignancy Surgical Research Unit (HEPSURU), Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Colm J. O'Rourke
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sophie Bull Nordkild
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Hepatic Malignancy Surgical Research Unit (HEPSURU), Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Jan-Michael Kugler
- Institute for Molecular and Cellular Medicine, University of Copenhagen, Panum Institute, Copenhagen, Denmark
| | - Susanne Dam Nielsen
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Viro-immunology Research Unit, Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
- Institute for Clinical Medicine, University of Copenhagen, Panum Institute, Copenhagen, Denmark
| | - Jesper B. Andersen
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans-Christian Pommergaard
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Hepatic Malignancy Surgical Research Unit (HEPSURU), Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
- Institute for Clinical Medicine, University of Copenhagen, Panum Institute, Copenhagen, Denmark
| |
Collapse
|
67
|
Li M, Gao X, Lin X, Zhang Y, Peng W, Sun T, Shu W, Shi Y, Guan Y, Xia X, Yi X, Li Y, Jia J. Analysis of germline-somatic mutational connections in colorectal cancer reveals differential tumorigenic patterns and a novel predictive marker for germline mutation carriers. Cancer Lett 2025; 620:217637. [PMID: 40118241 DOI: 10.1016/j.canlet.2025.217637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/23/2025]
Abstract
Colorectal cancer (CRC) genetic testing of regions beyond clinical guidelines has revealed a substantial number of likely pathogenic germline mutations (GMs). It remains largely undetermined whether and how these GMs, typically located in non-mismatch repair (non-MMR) genes, are associated with the tumorigenesis of CRC. This study aimed to identify CRC-predisposing GMs among 93 cancer susceptibility genes and investigate their potential influences on CRC somatic mutational features. We secondarily aimed to investigate whether somatic ERBB2 amplification contributes to identifying GM carriers. This study incorporated a total of 3,240 Chinese CRC patients and 10,588 control individuals. CRC patients were subjected to paired tumor-normal sequencing with a 1,021-gene panel. A case-control analysis was conducted to profile the GM-associated CRC risk. A comprehensive germline-somatic association analysis was performed among 2,405 patients, with key findings subsequently validated in an independent 835-patient cohort and the TCGA CRC cohort. The case-control results supported CRC-predisposing effects of GMs in certain homologous recombination repair (HRR) and DNA damage checkpoint factor (CPF) genes, such as BRCA1/2, RecQ helicase genes, ATM, and CHEK2. HRR GMs were associated with an increased copy number alteration burden, more TP53 clonal mutations, and a higher probability of carrying somatic ERBB2 amplification. CPF GMs were inferred to have synergistic effects with ARID1A and KDM6A somatic mutations in CRC tumorigenesis. Among patients with onset age ≥55 years, stable microsatellites, and no cancer family history, ERBB2 amplification was significantly predictive of GM carriers. Our findings elucidate different germline tumorigenic patterns not driven by deficient MMR. Somatic ERBB2 amplification in CRC can serve as an indicator for germline genetic testing when traditional risk features are absent.
Collapse
Affiliation(s)
- Mintao Li
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Xuan Gao
- Geneplus-Shenzhen Clinical Laboratory, Shenzhen, China
| | - Xiangchun Lin
- Department of Gastroenterology, Peking University International Hospital, Beijing, China
| | - Yan Zhang
- Geneplus-Beijing Institute, Beijing, China
| | - Wenying Peng
- The Second Department of Oncology, Yunnan Cancer Hospital & the Third Affiliated Hospital of Kunming Medical University & Yunnan Cancer Center, Kunming, China
| | - Tao Sun
- General Surgery Department, Peking University Third Hospital, Beijing, China
| | - Weiyang Shu
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Yanyan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | | | | | - Xin Yi
- Geneplus-Beijing Institute, Beijing, China.
| | - Yuan Li
- Department of Gastroenterology, Peking University International Hospital, Beijing, China; Department of Gastroenterology, Peking University Third Hospital, Beijing, China.
| | - Jinzhu Jia
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China; Center for Statistical Science, Peking University, Beijing, China.
| |
Collapse
|
68
|
Zheng X, Zhang X, Yu J, Zheng J. Pan-cancer analysis identifies EIPR1 as a potential prognostic and immunological biomarker for lung adenocarcinoma and its functional validation. Gene 2025; 954:149439. [PMID: 40154585 DOI: 10.1016/j.gene.2025.149439] [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: 11/18/2024] [Revised: 03/15/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND EARP and GARP complex-interacting protein 1 (EIPR1) may be a new oncogene in tumors, influencing the prognosis and invasion of cancer. However, a systematic analysis of the function of EIPR1 in various cancers remains vacant. Thus, we proceeded with a comprehensive analysis to ascertain the role of EIPR1 among various cancers. METHODS We explored EIPR1 expression in pan-cancer, and its association with clinical stage, survival, gene mutations and methylation by the TIMER 2.0, GEPIA2, cBioPortal, and UALCAN. The protein-protein interaction (PPI) network, immune infiltration, and immune checkpoint assessments of EIPR1 was performed using the STRING and SangerBox. The role of EIPR1 expression in lung adenocarcinoma (LUAD) was explored by the R software. The impact of EIPR1 expression on LUAD progression was studied through in vitro assays. RESULTS EIPR1 was overexpressed in most cancers and revealed as a potential prognostic biomarker in tumors, involving in tumorigenesis by affecting its methylation and gene mutations. The immune infiltration and immune checkpoints of tumors were related to the expression of EIPR1. Additionally, EIPR1 expression affected the survival, diagnosis, clinicopathological features, tumor microenvironment, and drug sensitivity of LUAD patients. Validation studies demonstrated that EIPR1 knockdown suppressed the malignant growth, invasion, and migration of LUAD cells. CONCLUSIONS This study delivers an extensive landscape for the oncogenesis and immunological characteristics of EIPR1, which reveals that EIPR1 may serve as a potential biological target for future prognosis and immune treatment in tumors, especially in LUAD.
Collapse
Affiliation(s)
- Xin Zheng
- Department of Diagnostic Pathology, Shandong Second Medical University, Weifang 261053, China
| | - Xiao Zhang
- Department of Ultrasound, Weifang People's Hospital, Weifang 261041, China
| | - Jie Yu
- Department of Diagnostic Pathology, Shandong Second Medical University, Weifang 261053, China
| | - Jie Zheng
- Department of Diagnostic Pathology, Shandong Second Medical University, Weifang 261053, China; Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
69
|
Zubareva EY, Senchukova MA, Saidler NV. Cytoplasmic and nuclear programmed death ligand 1 expression in peritumoral stromal cells in breast cancer: Prognostic and predictive value. World J Exp Med 2025; 15:102761. [DOI: 10.5493/wjem.v15.i2.102761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/21/2025] [Accepted: 02/21/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Breast cancer (BC) continues to occupy a leading position in terms of morbidity and mortality from malignant neoplasms among the female population. One of the promising markers associated with BC progression is programmed death ligand 1 (PD-L1). Previously, we investigated PD-L1 expression in BC via a new antibody against programmed cell death protein 1 ligand 1 (PDCD1 LG1) and reported that high PDCD1 LG1 expression in tumor cells is an independent factor for a high risk of regional metastasis in patients with BC. However, the prognostic significance of PDCD1 LG1 expression in BC stromal cells has not been adequately studied.
AIM To study the features of PDCD1 LG1 expression in BC stromal cells and its relationship with BC clinicopathological characteristics.
METHODS In a prospective single-center observational study, tumor samples from 148 patients with newly diagnosed BC were examined. The tumor sections were immunohistochemically stained with antibodies against PDCD1 LG1. In the tumor samples, the PDCD1 LG1-positive lymphocyte (PDCD1 LG1+ LF) score, presence of nuclear PDCD1 LG1 expression in the LFs, PDCD1 LG1 expression in polymorphic cell infiltrates (PDCD1 LG1+ polymorphic cell infiltrates [PCIs]), and cells of the fibroblastic stroma and endothelial cells of the tumor microvessels were assessed. Statistical analyses were performed using Statistica 10.0 software.
RESULTS A PDCD1 LG1+ LF score ≥ 3 was detected more often at stages N0 and N3 than at N1 and N2 (P = 0.03). Moderate and pronounced PDCD1 LG1+ PCIs and the presence of PDCD1 LG1+ fibroblastic stroma were associated with negative estrogen receptor status (P = 0.0008 and P = 0.03, respectively), human epidermal growth factor receptor 2-positive (HER2+) BC (P < 0.00001 and P = 0.0005), and luminal B HER2+, non-luminal HER2+ and triple-negative BC (P < 0.00001 and P = 0.004). The risk of metastasis to regional lymph nodes (RLNs) depend on lymphovascular invasion (LVI) and the PDCD1 LG1+ LF score. In the absence of LVI and a PDCD1 LG1+ LF score < 3 or ≥ 3, metastases in RLNs were absent in 66.6% and 93.9% of patients with BC, respectively. In the presence of LVI and a PDCD1 LG1+ LF score < 3 or ≥ 3, metastases in RLNs were detected in 82.6% and 92.7% of patients with BC, respectively.
CONCLUSION The results indicated that the combined assessment of the PDCD1 LG1+ LF score and LVI can improve the accuracy of predicting the risk of metastasis to RLNs in patients with BC.
Collapse
Affiliation(s)
- Evgeniya Yu Zubareva
- Department of Oncology, Orenburg State Medical University, Orenburg 460021, Orenburgskaya Oblast, Russia
| | - Marina A Senchukova
- Department of Oncology, Orenburg State Medical University, Orenburg 460021, Orenburgskaya Oblast, Russia
| | - Natalia V Saidler
- Department of Pathology, Orenburg Regional Cancer Clinic, Orenburg 460021, Orenburgskaya Oblast, Russia
| |
Collapse
|
70
|
Goloudina A, Le Chevalier F, Authié P, Charneau P, Majlessi L. Shared neoantigens for cancer immunotherapy. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200978. [PMID: 40256120 PMCID: PMC12008704 DOI: 10.1016/j.omton.2025.200978] [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] [Indexed: 04/22/2025]
Abstract
Exploration of neoantigens holds the potential to be productive in immuno-oncotherapy. Among tumor-specific antigens, neoantigens result from genetic instability that gives rise to non-synonymous somatic mutations, highly specific to tumor cells. In addition to point mutations, gene rearrangements, indels leading to frameshifts, chromosomal translocations or inversions that may lead to fusion proteins, alternative mRNA splicing, and integration of genetic material of oncogenic viruses into the host genome provide consistent sources of neoantigens that are absent in healthy tissues. Out of these alterations, 2%-3% may generate T cell neoepitopes, possibly detectable by TCRs. Neoantigens are absent in healthy tissues and are thus at low risk of triggering autoimmunity. In addition, the host lymphocytes have not been rendered tolerant toward them and it is possible to induce immune responses against them. Here, we overview the two categories of neoantigens, i.e., private and shared, and their use in immuno-oncotherapy in selected pre-clinical and clinical studies. The vast majority of commonly occurring tumor-specific mutations are cancer causing and are permanently expressed by all malignant tumor cells, preventing the latter from escaping vaccine-induced anti-neoantigen immunity. The use of public neoantigens combined with efficient vaccine platforms can provide non-personalized "off-the-shelf" therapeutic vaccine candidates for broad-spectrum immunotherapy purposes.
Collapse
Affiliation(s)
- Anastasia Goloudina
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Fabien Le Chevalier
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Authié
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Charneau
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Laleh Majlessi
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| |
Collapse
|
71
|
Xu S, Yuan X, Wang Y, Fu Z, Chen K, Cui Z, Xu L, Zhang H, Xia D, Wu Y. Bisphenols exposure at environmentally relevant dose promoted ovarian cancer progression and modulated tumor microenvironment through β-catenin/SPP1 axis. JOURNAL OF HAZARDOUS MATERIALS 2025; 490:137824. [PMID: 40054195 DOI: 10.1016/j.jhazmat.2025.137824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 02/18/2025] [Accepted: 03/01/2025] [Indexed: 04/16/2025]
Abstract
Bisphenol A (BPA) and its substitute, Bisphenol S (BPS) are typical endocrine-disrupting chemicals used in plastics, but their cancer-promoting effect has remained controversial. Here, we investigated the effects of environmentally relevant doses of BPA/BPS exposure on the tumor microenvironment (TME) in ovarian cancer. BPA exposure levels was exhibiting a declining trend and BPS showing an ascending trend in the female population by analyzing the NHANES data (2013-2016). Low doses of BPA/BPS both significantly promoted the migration and invasion of ovarian cancer cells in a dose-dependent manner by activating the Wnt/β-catenin signaling pathway, thereby facilitating the SPP1 gene transcription. Notably, low-dose BPA/BPS exposure stimulated ovarian cancer cells to secrete OPN protein (coded by the SPP1 gene), subsequently inducing the transformation of fibroblasts into cancer-associated fibroblasts (CAFs), which could reshape the TME of ovarian cancer. Two in-vivo experiments established with nude mice and SPP1-/- mice respectively, both confirmed that low-dose BPA/BPS exposure increased the incidence of tumor metastasis accompanied by CAF infiltration, while administration of OPN-neutralizing antibodies effectively blocked these effects. Our results indicated that exposure to either BPA or its substitute BPS could promote the release of secreted protein OPN via the β-catenin/SPP1 axis, ultimately modulating the TME and enhancing the progression of ovarian cancer, providing new evidence and potential intervention strategies for the toxicological assessment and management of bisphenols.
Collapse
Affiliation(s)
- Sinan Xu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyu Yuan
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuwei Wang
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiqin Fu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kelie Chen
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Gynecology and Obstetrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenyan Cui
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Leting Xu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Honghe Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Dajing Xia
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yihua Wu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
72
|
Yun Z, Liu Z, Sun Z, Yan X, Yang Q, Tian S, Li X, Hou L. Identification of potential drug targets for four site-specific cancers by integrating human plasma proteome with genome. J Pharm Biomed Anal 2025; 258:116731. [PMID: 39933395 DOI: 10.1016/j.jpba.2025.116731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/18/2025] [Accepted: 02/02/2025] [Indexed: 02/13/2025]
Abstract
Drug targets supported by genetic evidence with a several-fold higher probability of success in clinical trials. We performed a comprehensive proteome-wide Mendelian randomization (MR) analysis to identify causal proteins and potential therapeutic targets for four site-specific cancers. A total of 13,248 protein quantitative trait loci for 4853 plasma proteins were utilized for proteome-wide MR analysis. Identification of cancer causal proteins in the discovery cohort and further validation in the replication cohort. Colocalization, summary-data-based MR (SMR) analysis, and transcriptome‑wide association studies (TWAS) were performed to check the accuracy of the candidate proteins. Two-step MR analysis was used to explore the effects of plasma protein-mediated 248 modifiable factors on cancer. Phenome-wide MR (Phe-MR) analysis, druggability evaluation, and single-cell type expression analysis further assessed the potential of causal proteins. Combining the results of the meta-analysis of MR estimates from the two cohorts, 21, 2, 24 and 1 causal proteins were identified in breast, lung, prostate and stomach cancers, respectively. Evidence from colocalization, SMR analysis, and TWAS highlighted CD36, DNPH1, and PLXND1 as the most promising drug targets for breast cancer, and ZNF175 for prostate cancer. 1 new potential biomarker (PLXND1) for breast cancer, 2 new promising targets (RELL1, DEFB119) for lung cancer, and 8 new circulating biomarkers (ARFIP2, CCN6, CTRB2, HTR7, MRPL33, TNFRSF6B, VAMP5, ZNF175) for prostate cancer were firstly reported. Some plasma proteins may mediate the association of these cancers with other systemic diseases. Additionally, genetically predicted higher BMI and overweight may reduce breast cancer risk by altering CASP8, ADM, PLXND1, TNFRSF9, ULK3 and VSIG4 protein levels. Causal proteins of breast and prostate cancer were expressed predominantly on macrophages in cancerous tissues. This study genetically identified several cancer causal proteins which provided new perspectives for the understanding of the etiology and development of novel targeted drugs for cancer.
Collapse
Affiliation(s)
- Zhangjun Yun
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China; Beijing University of Chinese Medicine, Beijing 100700, China
| | - Zhu Liu
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China; Beijing University of Chinese Medicine, Beijing 100700, China
| | - Ziyi Sun
- Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xiang Yan
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China; Beijing University of Chinese Medicine, Beijing 100700, China
| | - Qianru Yang
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China; Beijing University of Chinese Medicine, Beijing 100700, China
| | - Shaodan Tian
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China.
| | - Xiao Li
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China.
| | - Li Hou
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China.
| |
Collapse
|
73
|
Akkus E, Karaoğlan BB, Kayaalp M, Turmuş U, Akyol C, Utkan G. Stage-specific characterization of "early-onset colorectal cancer": Localized and synchronous metastatic disease. Int J Cancer 2025; 156:2340-2351. [PMID: 39887374 PMCID: PMC12008821 DOI: 10.1002/ijc.35336] [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: 09/23/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025]
Abstract
Early-onset colorectal cancer (EOCRC) is an alarming entity worldwide. Yet, stage-specific characteristics and prognosis in localized and synchronous metastatic EOCRC are not well-defined. Two cohorts of CRC patients (localized and synchronous metastatic) were evaluated, defining EOCRC as the diagnosis <50 years old. Five hundred sixty-eight patients were included (n = 432 localized, 14.4% [n = 62] EOCRC and n = 136 synchronous metastatic, 20.6% [n = 28] EOCRC). 93.5% of localized and 96.5% of synchronous metastatic EOCRC patients were symptomatic at diagnosis. Among localized patients, female gender (58.1% vs. 40%, p = .008), perineural invasion (41.9% vs. 24.9%, p = .005), folinic acid, 5-fluorouracil, and oxaliplatin chemotherapy (45.2% vs. 25.2%, p = .003), and perioperative chemotherapy cycles (9.21 [± 3.10] vs. 7.98 [± 2.92], p = .006) were higher in EOCRC compared with ≥50-year. Median recurrence-free survival (RFS) and overall survival were not reached in either group (p = .234 and p = .831). Only RAS mutant status was associated with RFS (Hazard ratio: 7.09 [95% confidence interval (CI): 1.87-26.76], p < .001) in EOCRC. Among synchronous metastatic patients, urgent surgery (32.1% vs. 11.1%, p = .014) and local treatments (39.3% vs. 20.4%, p = .037) were more frequent in EOCRC. Median progression-free survival and overall survival in the EOCRC and ≥50 years were 8.07 months (95% CI: 5.03-12.97) vs. 10.03 months (95% CI, 8.40-13.10) (p = .450) and 18.57 months (95% CI, 13.33-43.03) vs. 19.83 months (95% CI, 16.07-27.30) (p = .833), respectively. Synchronous metastatic EOCRC more frequently underwent urgent surgery (32.1% vs. 8%, p = .008) and had RAS mutation (43.5% vs. 16.7%, p = .032) than localized EOCRC. This study suggests that localized and synchronous metastatic EOCRC patients may have different characteristics than average onset, without survival differences. Implementation of stage-specific characteristics into daily practice is necessary for decision-making processes in these young patients.
Collapse
Affiliation(s)
- Erman Akkus
- Department of Medical OncologyAnkara University Faculty of MedicineAnkaraTürkiye
- Ankara University Cancer Research InstituteAnkaraTürkiye
| | - Beliz Bahar Karaoğlan
- Department of Medical OncologyAnkara University Faculty of MedicineAnkaraTürkiye
- Ankara University Cancer Research InstituteAnkaraTürkiye
| | - Mehmet Kayaalp
- Department of Medical OncologyAnkara University Faculty of MedicineAnkaraTürkiye
- Ankara University Cancer Research InstituteAnkaraTürkiye
| | - Utkucan Turmuş
- Department of Internal MedicineAnkara University Faculty of MedicineAnkaraTürkiye
| | - Cihangir Akyol
- Department of SurgeryAnkara University Faculty of MedicineAnkaraTürkiye
| | - Güngör Utkan
- Department of Medical OncologyAnkara University Faculty of MedicineAnkaraTürkiye
- Ankara University Cancer Research InstituteAnkaraTürkiye
| |
Collapse
|
74
|
Bendinelli B, Danza G, Assedi M, Villanelli F, Vagnoni G, Pastore E, Saieva C, Caini S, Vignozzi L, Masala G. Circulating sex hormones and volumetric breast density: A prospective study in women from the EPIC Florence cohort. Int J Cancer 2025; 156:2294-2302. [PMID: 40145865 PMCID: PMC12008827 DOI: 10.1002/ijc.35321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 03/28/2025]
Abstract
Mammographic breast density (MBD) is a known risk factor for breast cancer. This study aimed to evaluate prospectively how circulating levels of sex hormones and sex hormones binding globulin (SHBG) are associated with volumetric percent density (VPD, %) and absolute breast dense volume (DV, cm3) measured after an average 13.5 years of follow up. We included 422 post- and 445 pre-menopausal women enrolled in the Florence section of the European Prospective Investigation into Cancer and Nutrition study. Blood levels of circulating sex hormones and SHBG were determined in serum samples collected at enrollment. The automated Volpara™ density software was used to obtain DV and VPD measures from the first available full-field digital mammography performed after enrollment. We carried out adjusted linear regression models for post-menopausal and pre-menopausal women, separately. Higher baseline blood levels of SHBG were associated with higher VPD both in post-menopausal (%Diff 17.2 III vs. I tertile, p-value 0.02, p-trend 0.02) and pre-menopausal women (%Diff 21.1 III vs. I tertile, p-value 0.004, p-trend 0.004). Among pre-menopausal women a modifying effect of parity emerged, with significant inverse associations of estradiol, free estradiol, estrone, and free testosterone with VPD in women with a lower number of births or higher age at every birth. DV was not associated with sex hormones or SHBG. A direct association between increasing serum level of SHBG and VPD was observed both in post- and pre-menopausal women. This association need to be further investigated in order to better understand the underlying mechanisms.
Collapse
Affiliation(s)
- Benedetta Bendinelli
- Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO)FlorenceItaly
| | - Giovanna Danza
- Department of Experimental Clinical and Biomedical Sciences "Mario Serio"University of FlorenceFlorenceItaly
| | - Melania Assedi
- Cancer Risk Factors and Lifestyle Epidemiology UnitInstitute for Cancer Research, Prevention and Clinical Network (ISPRO)FlorenceItaly
| | - Fabio Villanelli
- Department of Experimental Clinical and Biomedical Sciences "Mario Serio"University of FlorenceFlorenceItaly
| | - Giulia Vagnoni
- Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO)FlorenceItaly
| | - Elisa Pastore
- Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO)FlorenceItaly
| | - Calogero Saieva
- Cancer Risk Factors and Lifestyle Epidemiology UnitInstitute for Cancer Research, Prevention and Clinical Network (ISPRO)FlorenceItaly
| | - Saverio Caini
- Cancer Risk Factors and Lifestyle Epidemiology UnitInstitute for Cancer Research, Prevention and Clinical Network (ISPRO)FlorenceItaly
| | - Linda Vignozzi
- Department of Experimental Clinical and Biomedical Sciences "Mario Serio"University of FlorenceFlorenceItaly
- Centro Regionale di Coordinamento Salute e Medicina di Genere (CISMEG)Careggi University HospitalFlorenceItaly
| | - Giovanna Masala
- Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO)FlorenceItaly
| |
Collapse
|
75
|
Harper DM, Paczos T, Ridder R, Huh WK. p16/ki-67 dual stain triage of individuals positive for HPV to detect cervical precancerous lesions. Int J Cancer 2025; 156:2257-2264. [PMID: 39901857 PMCID: PMC12008826 DOI: 10.1002/ijc.35353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 02/05/2025]
Abstract
p16/Ki-67 dual stain is a biomarker-based test that can identify oncogenic transformation in cervical cells with higher sensitivity than cervical cytology, using the same samples taken for human papillomavirus (HPV) testing and liquid-based cytology. Dual stain is approved by the US Food and Drug Administration (FDA) for triage of women with positive results by primary HPV testing or by HPV/cytology co-testing and has recently been incorporated into management guidelines. In this review, we summarize the data showing the utility of dual stain in detecting precancers, reducing the number of unnecessary colposcopies, and reassuring women who test negative. We also discuss the implications of dual stain for future treatment practice and health economics.
Collapse
Affiliation(s)
| | | | | | - Warner K. Huh
- University of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
76
|
Ganbold E, Kim NY, Kim YM, Sharma PK, Lee DN, Oh B, Kim HS, Song J, Lee B, Kim ES, Shin YK, Park JS, Kim ST. Reagentless aptamer based on the ultrasensitive and fast response electrochemical capacitive biosensor for EGFR detection in non-small cell lung cancer. Biosens Bioelectron 2025; 278:117319. [PMID: 40112520 DOI: 10.1016/j.bios.2025.117319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/16/2025] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
Non-small cell lung cancer (NSCLC) is still the leading cause of lung cancer-related deaths globally, affecting both men and women. Mutations in the epidermal growth factor receptor (EGFR) are most common among patients with NSCLC, especially Asian patients. Here, we introduce an electrochemical capacitive biosensor for the early detection of NSCLC through specific identification of EGFR. A novel and reagentless EGFR aptamer was designed using the systematic evolution of ligands by exponential enrichment (SELEX) process and immobilized on a chromium (Cr)/gold (Au) electrode, with capacitance signals used for detection. The biosensor employs an interdigitated capacitor electrode (IDCE) functionalized with 3-mercaptopropionic acid (MPA), enhancing EGFR aptamer immobilization, while 6-mercapto-1-hexanol (MCH) was used for effective blocking to ensure robust and high-affinity binding to target analytes. The IDCE capacitive biosensor achieved real-time rapid detection within 3 s and demonstrated a detection limit of 0.005 ng/mL for the EGFR peptide, with a dynamic range of 10-11-10-7 ng/mL. Furthermore, the specific EGFR aptamer-immobilized IDCE biosensor was found to be regenerable and reusable up to five times using deionized water. This biosensor offers a rapid, label-free, and highly selective approach for early-stage EGFR detection in NSCLC. Its portability and scalability make it a promising tool for point-of-care diagnostic applications in biomedicine, potentially advancing the field of cancer diagnostics.
Collapse
Affiliation(s)
- Enkhzaya Ganbold
- RFIC Bio Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; Department of Electronics Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; NDAC Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea
| | - Nam Young Kim
- RFIC Bio Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; Department of Electronics Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; NDAC Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yu Mi Kim
- RFIC Bio Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; Neuroscience Research Institute, JnPharma Inc. Healthcare Innovation Park, Seoul National University College of Medicine, Goomi-ro, Bundanggu, Seongnam City, Gyeonggi-do 13605, Republic of Korea
| | - Parshant Kumar Sharma
- RFIC Bio Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; Department of Electronics Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea
| | - Do Nam Lee
- RFIC Bio Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; Ingenium College of Liberal Arts (Chemistry), Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea
| | - Byeolnim Oh
- RFIC Bio Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; Department of Electronics Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea
| | - Hyun Soo Kim
- RFIC Bio Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; Department of Electronics Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea
| | - Junghan Song
- RFIC Bio Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; Department of Laboratory Medicine, Seoul National University Bundang Hospital, Goomi-ro, Bundanggu, Seongnam City, Gyeonggi-do 13605, Republic of Korea
| | - Byungheon Lee
- RFIC Bio Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; Department of Biochemistry, School of Medicine, Kyungpook National University 680 Guckchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
| | - Eun-Seong Kim
- RFIC Bio Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| | - Young Kee Shin
- RFIC Bio Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| | - Jeong Su Park
- RFIC Bio Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; Department of Laboratory Medicine, Seoul National University Bundang Hospital, Goomi-ro, Bundanggu, Seongnam City, Gyeonggi-do 13605, Republic of Korea
| | - Sang Tae Kim
- RFIC Bio Centre, Kwangwoon University, 20 Kwangwoon-ro, Nowon-Gu, Seoul 01897, Republic of Korea; Neuroscience Research Institute, JnPharma Inc. Healthcare Innovation Park, Seoul National University College of Medicine, Goomi-ro, Bundanggu, Seongnam City, Gyeonggi-do 13605, Republic of Korea.
| |
Collapse
|
77
|
Wang A, Mizejewski GJ, Zhang C. Growth inhibitory peptides: a potential novel therapeutic approach to cancer treatment. Eur J Pharmacol 2025; 996:177554. [PMID: 40147579 DOI: 10.1016/j.ejphar.2025.177554] [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: 01/02/2025] [Revised: 03/02/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Cancer remains a major global public health concern, with considerable interest in exploring biological molecules for cancer treatment and prevention. Growth inhibitory peptide (GIP), a promising new class of biological therapeutics, has drawn attention for its distinct anti-tumor properties. Derived from human alpha-fetoprotein (HAFP), this synthetic 34-amino-acid peptide has demonstrated substantial anti-tumor effects across various cancer cell lines, effectively inhibiting tumor cell proliferation, migration, and metastasis. Studies reveal that GIP mediates its effects through a range of mechanisms, including interactions with G protein-coupled receptors (GPCRs), anti-cell adhesion activities, inhibition of cell spreading and metastatic processes, morphological alterations, platelet aggregation inhibition, immune enhancement, cell membrane disruption, ion channel blockade, and cell cycle arrest. While GIP has exhibited promising anti-tumor activity in both in vitro and in vivo models, further investigation is essential to advance its development as a therapeutic drug, particularly regarding pharmacokinetics, safety profiles, storage stability, and clinical efficacy.
Collapse
Affiliation(s)
- Aixin Wang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - G J Mizejewski
- Division of Translational Medicine, Molecular Diagnostics Laboratory, Wadsworth Center, New York State Department of Health Biggs Laboratory, Empire State Plaza Albany, NY 12237, USA
| | - Chao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| |
Collapse
|
78
|
Li G, Chen W, Liu D, Tang S. Recent advances in medicinal chemistry strategies for the development of METTL3 inhibitors. Eur J Med Chem 2025; 290:117560. [PMID: 40147343 DOI: 10.1016/j.ejmech.2025.117560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
N6-methyladenosine (m6A), the most abundant RNA modification in eukaryotic cells, exerts a critical influence on RNA function and gene expression. It has attracted considerable attention within the rapidly evolving field of epitranscriptomics. METTL3 is a key enzyme for m6A modification and is essential for maintaining normal m6A levels. High expression of METTL3 is closely associated with various cancers, including gastric cancer, liver cancer, and leukemia. Inhibiting METTL3 has shown potential in slowing cancer progression, thereby driving the development of METTL3 inhibitors. In this work, we summarize recent advancements in the development of METTL3 inhibitor, with a focus on medicinal chemistry strategies employed during discovery and optimization phases. We explore the application of structure-activity relationship (SAR) studies and protein-targeted degradation techniques, while addressing key challenges associated with their characterization and clinical translation. This review underscores the therapeutic potential of METTL3 inhibitors in modulating epitranscriptomic pathways and aims to offer perspectives for future research in this rapidly evolving field.
Collapse
Affiliation(s)
- Gengwu Li
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Wei Chen
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Shibing Tang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
79
|
Zhang Y, Luo Z, Jiang Y, Zheng L, Ma L, Zheng Y, Zou M, Kong L, Wang X. Discovery of dual CDK4/6 and BRD4 inhibitor as apoptosis and autophagy inducers against NSCLC in vitro and in vivo. Eur J Med Chem 2025; 290:117495. [PMID: 40101452 DOI: 10.1016/j.ejmech.2025.117495] [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: 01/21/2025] [Revised: 02/26/2025] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
Target of cyclin dependent kinase (CDK) by inhibitors has demonstrated promising potential as a therapeutic agent for cancer. However, the efficacy of monotherapy on tumors is limited and there is an urgent need for combination therapy with other inhibitors. It has been reported that restoring bromodomain-containing protein 4 (BRD4) resensitivity to tumor cells by inhibiting CDK4/6 is a potential therapeutic strategy. In this study, we present the design and optimization of dual CDK4/6 and BRD4 inhibitors, among which B15 exhibited potent and selective inhibition of both targets in vitro, and significant antiproliferative effects in non-small cell lung cancer (NSCLC) cells. Importantly, it also showed good pharmacokinetic properties in rats, meanwhile, B15 effectively inhibited tumor growth in vivo (TGI = 85.3 %) without causing significant toxicity. Overall, our results introduce a promising strategy of dual CDK4/6 and BRD4 inhibitors for the treatment of NSCLC.
Collapse
Affiliation(s)
- Yonglei Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Zhongwen Luo
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yuhan Jiang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Long Zheng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Liangliang Ma
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yiwei Zheng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Meiting Zou
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China.
| | - Xiaobing Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China.
| |
Collapse
|
80
|
Pejman Sani M, Mohseni S, Samimi H, Nasiri S, Fallahi B, Mohajeri-Tehrani M, Tavangar SM, Naderi M, Shirzad N, Larijani B, Sajjadi-Jazi SM, Roshandel G, Haghpanah V. Continued rise in the incidence of thyroid cancer in Iran: true increase or overdiagnosis? J Diabetes Metab Disord 2025; 24:68. [PMID: 39980844 PMCID: PMC11836260 DOI: 10.1007/s40200-025-01581-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 02/02/2025] [Indexed: 02/22/2025]
Abstract
Objectives Thyroid cancer (TC) is commonly recognized as the most prevalent type of malignancy affecting the endocrine system. This study aimed to assess the incidence of TC and its trends in the Iranian population. Methods The incidence rate of TC in Iran was determined using data from the Iranian National Population-based Cancer Registry (INPCR). The INPCR registered all new cancer cases through various diagnostic methods, including pathology reports, clinical and paraclinical data, and death registry reports. Results From 2014 to 2018, a total of 27,530 cases of TC were recorded. Among these cases, 21,932 (79.7%) were female, and 5,598 (20.3%) were male. The age-standardized incidence rate (ASR) of TC was 6.17 (95% confidence interval [CI]: 6.09-6.25) per 100,000 person-years, showing an upward trend from 4.61 (95% CI: 4.45-4.77) per 100,000 population in 2014 to 8.17 (95% CI: 7.97-8.37) in 2018. The ASR of TC in women was nearly 3.7 times higher than that in men (9.79 vs. 2.59 per 100,000 person-years). The ASR of TC was highest in younger age groups among women (40-50 years) compared to men, who had higher rates in older age groups (65-75 years). Papillary thyroid carcinoma (PTC), including its follicular variant, was the predominant histological type of TC in the Iranian population, accounting for 82.19% (n = 22,627) of cases, followed by follicular thyroid carcinoma (FTC) (n = 859; 3.12%). Conclusions Our data suggest that thyroid cancer rate has increased in Iran though comprehending the underlying reasons for this phenomenon requires further research.
Collapse
Affiliation(s)
- Mahnaz Pejman Sani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahrzad Mohseni
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hilda Samimi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirzad Nasiri
- Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Fallahi
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Mohajeri-Tehrani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Department of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Naderi
- Digestive Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nooshin Shirzad
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center (EMRC), Tehran University of Medical Sciences, Vali-Asr Hospital, Imam Khomeini Complex Hospital, Keshavarz Boulevard, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayed Mahmoud Sajjadi-Jazi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Roshandel
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Vahid Haghpanah
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
81
|
Kumar ARK, Low J, Lim J, Myint B, Sun X, Wu L, Cheng HS, Yip S, Ming Cheng CZ, Manoharan T, Quek YJ, Shou Y, Tian JS, Ng YY, Gascoigne NRJ, Tan NS, Sugimura R, Chia G, Sze Cheung AM, Yawata M, Tay A. Non-viral, high throughput genetic engineering of primary immune cells using nanostraw-mediated transfection. Biomaterials 2025; 317:123079. [PMID: 39842078 DOI: 10.1016/j.biomaterials.2024.123079] [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: 11/14/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025]
Abstract
Transfection of proteins, mRNA, and chimeric antigen receptor (CAR) transgenes into immune cells remains a critical bottleneck in cell manufacturing. Current methods, such as viruses and bulk electroporation, are hampered by low transfection efficiency, unintended transgene integration, and significant cell perturbation. The Nanostraw Electro-actuated Transfection (NExT) technology offers a solution by using high aspect-ratio nanostraws and localized electric fields to precisely deliver biomolecules into cells with minimal disruption. We demonstrate that NExT can deliver proteins, polysaccharides, and mRNA into primary human CD8+ and CD4+ T cells, and achieve CRISPR/Cas9 gene knockout of CXCR4 and TRAC in CD8+ T cells. We showcase NExT's versatility across a range of primary human immune cells, including CD4+ T cells, γδ-T cells, dendritic cells, NK cells, Treg cells, macrophages, and neutrophils. Finally, we developed a scalable, high-throughput multiwell NExT system capable of transfecting over 14 million cells and delivering diverse cargoes into multiple cell types from various donors simultaneously. This technology holds promise for streamlining high-throughput screening of allogeneic donors and reducing optimization costs for large-scale CAR-immune cell transfection.
Collapse
Affiliation(s)
- Arun R K Kumar
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Jessalyn Low
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Jet Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Ba Myint
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Xinhong Sun
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Ling Wu
- Immunology Translational Research Programme and Department of Microbiology and Immunology, National University of Singapore, Singapore, 117545, Singapore
| | - Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Sophronia Yip
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong Kong, Hong Kong SAR, China; Centre for Translational Stem Cell Biology, Hong Kong SAR, China
| | - Cyrus Zai Ming Cheng
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Thamizhanban Manoharan
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Ying Jie Quek
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Johann Shane Tian
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Yu Yang Ng
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Nicholas R J Gascoigne
- Immunology Translational Research Programme and Department of Microbiology and Immunology, National University of Singapore, Singapore, 117545, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Rio Sugimura
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong Kong, Hong Kong SAR, China; Centre for Translational Stem Cell Biology, Hong Kong SAR, China
| | - Gloryn Chia
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Alice Man Sze Cheung
- Department of Haematology, Singapore General Hospital, Singapore, 169608, Singapore; SingHealth Duke-NUS Medicine Academic Clinical Program, Duke-NUS Medical School, Singapore, 168753, Singapore
| | - Makoto Yawata
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 119077, Singapore
| | - Andy Tay
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore; NUS Tissue Engineering Program, National University of Singapore, Singapore, 117510, Singapore.
| |
Collapse
|
82
|
Tan T, Yuan S, Chu W, Jiang J, Chen M, Xia Q, Wang J. Low-dose anlotinib plus immune checkpoint inhibitors offers better efficacy and safety in advanced non-small cell lung cancer treatment. Anticancer Drugs 2025; 36:408-414. [PMID: 39992081 PMCID: PMC11969353 DOI: 10.1097/cad.0000000000001701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 01/13/2025] [Indexed: 02/25/2025]
Abstract
The combination of anlotinib with immune checkpoint inhibitors (ICIs) has become a common treatment modality in clinical practice. However, the optimal dose of anlotinib to use remains unclear. We collected patients with advanced non-small cell lung cancer (NSCLC) who received programmed cell death-1 blockade combined with different dose of anlotinib as second-line or later line therapy. Subsequently, the efficacy and safety of the combination therapy as well as subgroup analyses of different doses of anlotinib were analyzed. Cox regression was performed to analyze significant factors correlated with progression-free survival (PFS) and overall survival (OS). A total of 50 eligible patients with NSCLC who received anlotinib combined with ICIs therapy were included, of which 27 received low-dose anlotinib (8 mg), and 23 were administered high-dose anlotinib (12 mg). The median PFS (mPFS) and the median OS (mOS) for all patients were 8.3 months [95% confidence interval (CI): 6.3-10.3] and 17.6 months (95% CI: 16.5-18.7), respectively. Subgroup analyses showed that patients treated with 8 mg of anlotinib plus ICIs had significantly longer mPFS than those treated with 12 mg of anlotinib plus ICIs (8.7 vs 6.7 months, P = 0.016). The overall incidence of adverse events was 68.0%, and the most common adverse events of all grades were hypertension. Meanwhile, the incidence of adverse events was higher for 12 mg of anlotinib plus ICIs than that of 8 mg of anlotinib plus ICIs (82.6 vs 55.6%, P = 0.041). Low-dose anlotinib in combination with ICIs for advanced NSCLC may be an effective and well-tolerated option.
Collapse
Affiliation(s)
- Tingfei Tan
- Pharmacy Center, Hefei Cancer Hospital, Chinese Academy of Sciences
| | - Siyu Yuan
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University
| | - Weiwei Chu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiemei Jiang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Meiling Chen
- Pharmacy Center, Hefei Cancer Hospital, Chinese Academy of Sciences
| | - Quan Xia
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Junping Wang
- Pharmacy Center, Hefei Cancer Hospital, Chinese Academy of Sciences
| |
Collapse
|
83
|
Peng W, Liang J, Qian X, Li M, Nie M, Chen B. IGF2BP1/AIFM2 axis regulates ferroptosis and glycolysis to drive hepatocellular carcinoma progression. Cell Signal 2025; 130:111660. [PMID: 39971223 DOI: 10.1016/j.cellsig.2025.111660] [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: 01/24/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is aggressive liver tumor that is the third leading cause of cancer death. Ferroptosis and glycolysis play key roles in HCC progression. Apoptosis-inducing factor mitochondria-associated 2 (AIFM2) in involved in regulating ferroptosis and glycolysis in cancers, but its role in HCC remains unclear. This research explored the function of AIFM2 in HCC. METHODS AIFM2 expression in HCC tissues was evaluated using the UALCAN and GEPIA databases, as well as RT-qPCR. Kaplan-Meier survival analysis analyzed the correlation between AIFM2 and the prognosis of HCC patients. EdU and transwell assays were utilized to examine HCC cell proliferation, migration, and invasion. Ferroptosis markers were analyzed by measuring iron levels, ROS production (DCFH-DA assay), and oxidative stress indicators (SOD, MDA, and GSH). Glycolytic activity was assessed through glucose uptake, lactate production, and ATP levels. m6A modification on AIFM2 mRNA was confirmed by MeRIP assay, and mRNA stability was evaluated with Actinomycin D treatment. Tumor growth and metastasis were studied in xenograft and lung metastasis models. RESULTS UALCAN analysis showed that AIFM2 was significantly upregulated in HCC tissues, which correlated with poor survival rates of HCC patients. IGF2BP1 was also highly expressed in HCC tissues and positively correlated with AIFM2 levels in HCC tissues. Functionally, AIFM2 knockdown suppressed glycolysis and enhanced ferroptosis, while its overexpression had opposite effects. IGF2BP1 was found to stabilize AIFM2 mRNA via m6A modification, promoting AIFM2 expression. IGF2BP1 knockdown reduced glycolysis, proliferation, and invasion while promoting ferroptosis, while AIFM2 overexpression could reverse this effect. In vivo, IGF2BP1 or AIFM2 silencing significantly suppressed tumor growth and metastasis. CONCLUSION IGF2BP1 stabilized AIFM2 mRNA to regulate ferroptosis and glycolysis and promoted HCC progression.
Collapse
Affiliation(s)
- Wei Peng
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Jie Liang
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Xuanlv Qian
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Mingwang Li
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Ming Nie
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Bin Chen
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China.
| |
Collapse
|
84
|
Jia P, Tu J, Shen H, Jiang Y, Zhang Q, Xue W, Liu M, Liu J, Miao Y, Ouyang R, Zhou S. Defect-engineered magnetic bismuth nanomedicine for dual-modal imaging and synergistic lung tumor therapy. Mater Today Bio 2025; 32:101680. [PMID: 40225134 PMCID: PMC11987673 DOI: 10.1016/j.mtbio.2025.101680] [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: 11/23/2024] [Revised: 02/28/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
Bismuth sulfide (Bi2S3) nanomaterials are recognized for their potential in tumor therapy due to their narrow bandgap and low toxicity. However, limited photothermal conversion efficiency (PCE) and low carrier density restrict their broader application in photothermal cancer treatment. To address these challenges, we designed defect-engineered, magnetic-targeting Bi2S3-based photothermal nanoparticles, Fe3O4@Au@Bi2S3 nanorugbys (Fe3O4@Au@Bi2S3 NRs). These nanoparticles were developed using a layer-by-layer encapsulation strategy with sulfur vacancies (Vs) and Bi antisite defects (Bi replacing S, Bis), enhancing electron trapping and recombination to boost the near-infrared (NIR) response. The PCE of Fe3O4@Au@Bi2S3 NRs reached 44.34 %, which significantly improved their efficacy in photothermal treatment for lung tumors. Moreover, the polyvinylpyrrolidone (PVP) coating on the nanoparticles enabled efficient loading and pH-responsive release of doxorubicin hydrochloride (DOX), facilitating synergistic chemo-photothermal therapy. When exposed to an external magnetic field, the nanoparticles demonstrated strong magnetic targeting and enhanced computed tomography (CT) imaging capabilities, improving tumor treatment accuracy. Both in vitro and in vivo studies showed that this combined therapy effectively induced cancer cell apoptosis and inhibited tumor proliferation, showcasing outstanding anti-tumor performance. This study provides a promising strategy for enhancing chemo-photothermal therapy through defect-engineered, magnetic-targeted Fe3O4@Au@Bi2S3 nanoparticles, holding significant potential for clinical applications in tumor therapy.
Collapse
Affiliation(s)
- Pengpeng Jia
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
- USST-UH International Joint Laboratory for Tumor Diagnosis and Energy Treatment, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Jie Tu
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
- USST-UH International Joint Laboratory for Tumor Diagnosis and Energy Treatment, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Hongyu Shen
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
- USST-UH International Joint Laboratory for Tumor Diagnosis and Energy Treatment, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuqin Jiang
- Collaborative Innovation Centre of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Engineering Research Centre of Chiral Hydroxyl Pharmaceutical, Henan Engineering Laboratory of Chemical Pharmaceutical and Biomedical Materials, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, 453007, China
| | - Qiupeng Zhang
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
- USST-UH International Joint Laboratory for Tumor Diagnosis and Energy Treatment, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Weixian Xue
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
- USST-UH International Joint Laboratory for Tumor Diagnosis and Energy Treatment, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Meitong Liu
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
- USST-UH International Joint Laboratory for Tumor Diagnosis and Energy Treatment, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Jianbo Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Yuqing Miao
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
- USST-UH International Joint Laboratory for Tumor Diagnosis and Energy Treatment, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Ruizhuo Ouyang
- Institute of Bismuth and Rhenium Science, University of Shanghai for Science and Technology, Shanghai, 200093, China
- USST-UH International Joint Laboratory for Tumor Diagnosis and Energy Treatment, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Shuang Zhou
- Cancer Institute, Tongji University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
85
|
Baandrup L, Kjær SK, Jacobsen Ó, Bzorek M, Eriksen TT, Larsen LG, Fiehn AMK. Development of a digital algorithm for assessing tumor-stroma ratio, tumor budding and tumor infiltrating lymphocytes in vulvar squamous cell carcinomas. Ann Diagn Pathol 2025; 76:152462. [PMID: 40048885 DOI: 10.1016/j.anndiagpath.2025.152462] [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: 01/10/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/23/2025]
Abstract
Tumor-stroma ratio (TSR), tumor budding (TB), and tumor-infiltrating lymphocytes (TILs) are prognostic markers in some cancers but with unknown significance in vulvar squamous cell carcinoma (VSCC). This pilot study primarily aimed to develop a digital method for evaluating TSR, TB and TILs in VSCC and secondarily to investigate variation in these factors by p16 status. An independent training set stained with CD3/cytokeratin and CD8/cytokeratin was used to develop a deep learning-based Application Protocol Package (APP) segmenting tissue into background, epithelium, or stroma. TSR was defined as percentage of tumor epithelium relative to total tumor area, and tumor buds were defined as clusters of 1-4 tumor cells. A second APP quantified CD3+ and CD8+ lymphocytes in the intraepithelial and stromal compartments, respectively. The digital algorithms were applied to the study cohort of 41 VSCC cases, achieving satisfactory performance without manual corrections. TSR ranged between 33 and 91% with median of 64%, and median number of buds was 4 (range: 0-48) buds/mm2. Median density and range of CD3+ lymphocytes were 222 (13-2320) cells/mm2 in the intraepithelial and 1978 (397-6683) cells/mm2 in the stromal compartment, respectively. CD8+ lymphocyte counts were lower. There was a tendency towards lower TSR and higher number of buds in p16-negative compared with p16-positive VSCC. Finally, automated measures were compared with manual evaluations showing high concordance. The developed automated method provided precise and objective measurements of TSR, TB and TILs. The algorithms should be validated in a larger cohort and correlated with clinicopathological characteristics and prognosis to determine their clinical relevance.
Collapse
Affiliation(s)
- Louise Baandrup
- Department of Pathology, Zealand University Hospital, Denmark; Unit of Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark.
| | - Susanne K Kjær
- Unit of Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark; Department of Gynecology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Óli Jacobsen
- Department of Pathology, Zealand University Hospital, Denmark
| | - Michael Bzorek
- Department of Pathology, Zealand University Hospital, Denmark
| | | | | | - Anne-Marie Kanstrup Fiehn
- Department of Pathology, Zealand University Hospital, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark
| |
Collapse
|
86
|
Inoue A, Kagawa Y, Nishizawa Y, Komori T, Hashimoto M, Ozato Y, Morimoto Y, Shimamoto S, Miyazaki Y, Tomokuni A, Motoori M, Fujitani K. Outcomes of neoadjuvant chemoradiotherapy for locally advanced rectal cancer under non‑smoking conditions confirmed by measuring expiratory CO levels: An observational study. Oncol Lett 2025; 29:266. [PMID: 40235682 PMCID: PMC11997641 DOI: 10.3892/ol.2025.15012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
The outcomes of neoadjuvant chemoradiotherapy (CRT) under non-smoking conditions in patients with locally advanced rectal cancer (LARC) remain unclear. The aim of the present study was to evaluate the outcomes in patients with LARC who underwent neoadjuvant CRT under non-smoking conditions, followed by total mesorectal excision (TME). To this end, the medical records of 28 patients treated with CRT and surgery for LARC between January 2014 and December 2019 were retrospectively analyzed. Smoking cessation was monitored by measuring carbon monoxide (CO) levels using a Smokerlyzer. Survival outcomes and clinicopathological factors associated with pathological complete response (pCR) were investigated. The median age was 66 (45-89) years, and 20 (71.4%) patients were male. A total of 16 (57.1%) patients were diagnosed with clinical stage III LARC. Seven patients smoked at diagnosis, with an average expiratory CO level of 8 (8-30) ppm. These patients ceased smoking and maintained exhaled CO levels <3 ppm before treatment. All patients successfully underwent CRT and TME. No major postoperative complications occurred. pCR was achieved in 8/28 patients (28.6%) and the 5-year recurrence-free and overall survival rates were 78.0% [95% confidence interval (CI), 57.4-89.5] and 85.7% (95% CI, 66.3-94.4), respectively. The median follow-up period was 60.1 (range, 5.6-114.6) months. Survival did not significantly differ between smokers and non-smokers at diagnosis. Clinically negative N stage (hazard ratio: 18.9; 95% CI, 1.63-218; P=0.0187) was significantly associated with pCR. In conclusion, neoadjuvant CRT under non-smoking conditions, as confirmed by measuring expiratory CO levels, followed by TME yields favorable pCR rates and survival outcomes in patients with LARC.
Collapse
Affiliation(s)
- Akira Inoue
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Osaka 558-8558, Japan
| | - Yoshinori Kagawa
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Osaka 558-8558, Japan
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Osaka 540-0008, Japan
| | - Yujiro Nishizawa
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Osaka 558-8558, Japan
| | - Takamichi Komori
- Department of Surgery, Hyogo Prefectural Nishinomiya Hospital, Nishinomiya, Hyogo 662-0918, Japan
| | - Masahiro Hashimoto
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Osaka 558-8558, Japan
| | - Yuki Ozato
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Osaka 558-8558, Japan
| | - Yoshihiro Morimoto
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Osaka 558-8558, Japan
| | - Shigetoshi Shimamoto
- Department of Radiation Oncology, Osaka General Medical Center, Osaka, Osaka 558-8558, Japan
| | - Yasuhiro Miyazaki
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Osaka 558-8558, Japan
| | - Akira Tomokuni
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Osaka 558-8558, Japan
| | - Masaaki Motoori
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Osaka 558-8558, Japan
| | - Kazumasa Fujitani
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Osaka 558-8558, Japan
| |
Collapse
|
87
|
Wen J, Cao X, Zhou B, Yang F, Wang X, Li Y, Zhao X, Mei J, Zhu W, Sun L, Huang F, Wang M. GC-MSCs transcriptionally upregulate SALL4 in gastric cancer through miR-4669/TIMP3/β-catenin signaling. Cell Signal 2025; 130:111668. [PMID: 39965736 DOI: 10.1016/j.cellsig.2025.111668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/07/2024] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUNDS Gastric cancer-associated mesenchymal stem cells (GC-MSCs) as integral components of the tumor microenvironment potentiate gastric cancer growth and metastasis. SALL4 is aberrantly upregulated in gastric cancer and pivotal for malignant progression. Whether GC-MSCs is responsible for SALL4 upregulation and the underlying mechanisms remains elusive. METHODS Cancer growth and metastasis capacities were assessed by cell colony formation assay, transwell assay, and epithelial-mesenchymal transition protein detection in vitro as well as subcutaneous xenograft and peritoneal metastasis models in vivo. SALL4 was measured by qPCR, western blot and immunohistochemistry staining. Gain- and loss-functional analysis were performed for miRNA and target gene. β-catenin signaling was assessed by immunofluorescence staining and Top/FopFlash luciferase assay. Transcriptional regulation was conducted using chemicals, luciferase reporter and ChIP assay. Clinical tissues and TCGA-STAD database were included for expression profile, correlation and clinical relevance analysis. RESULTS GC-MSCs promoted gastric cancer growth and metastasis along with elevation of SALL4 and miR-4669 in cancer cells and tissues. Overexpression of miR-4669 mimicked GC-MSC effects, while miR-4669 knockdown eliminated their oncogenic roles. TIMP3 was identified as a target of miR-4669 and mediated its functions. TIMP3 overexpression counteracted GC-MSC-induced cancer progression and SALL4 expression. GC-MSCs activated SALL4 transcription through the miR-4669/TIMP3/β-catenin pathway. The regulatory axis was aberrantly expressed in gastric cancer tissues, correlated with each other in certain cancer tissues and associated with lymph node metastasis. CONCLUSIONS GC-MSCs transcriptionally upregulate SALL4 to facilitate gastric cancer cell growth and metastasis via miR-4669/TIMP3/β-catenin pathway, highlighting the crucial role of GC-MSCs in the aberrant upregulation of SALL4.
Collapse
Affiliation(s)
- Jing Wen
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Xiaoli Cao
- Department of Laboratory Medicine, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu Province 226321, China
| | - Baocheng Zhou
- Department of Medical Laboratory, Lianyungang Maternal and Child Health Hospital, Lianyungang, Jiangsu Province 222000, China
| | - Fang Yang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Xiang Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Yuanyuan Li
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Xinlan Zhao
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Jingyu Mei
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Wei Zhu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China
| | - Li Sun
- Department of Clinical Laboratory, Kunshan First People's Hospital, Affiliated to Jiangsu University, Kunshan 215300, China
| | - Feng Huang
- Department of Clinical Laboratory, Kunshan First People's Hospital, Affiliated to Jiangsu University, Kunshan 215300, China; Department of Clinical Laboratory, Maternal and Child Health Care Hospital of Kunshan, Suzhou, Jiangsu Province, China
| | - Mei Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, China.
| |
Collapse
|
88
|
Qi M, Zhang K, Zhang X, Zhu Y, Cai B, Wang C, Zhao G, Zhang D, Zhang J. Arginine tagged liposomal carrier for the delivery of celastrol for ferroptosis-induced hepatocellular carcinoma therapy. Colloids Surf B Biointerfaces 2025; 250:114546. [PMID: 39919344 DOI: 10.1016/j.colsurfb.2025.114546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/21/2025] [Accepted: 01/31/2025] [Indexed: 02/09/2025]
Abstract
Hepatocellular carcinoma (HCC) is a predominant malignant liver tumor that cannot be efficiently treated because of poor response, toxicity, and drug resistance. Ferroptosis is an iron-dependent way of cell death associated with abnormal intracellular lipid metabolism. Celastrol (Cel) has the ability to inhibit the progression of HCC by regulating multiple signaling pathways and induce ferroptosis. However, Cel exists the limitations of low water solubility, low oral bioavailability, and high organ toxicity. Cel was encapsulated in polyethylene glycol-based liposomes modified with L-arginine (Cel@Lip-Arg). Cel@Lip-Arg has a uniform size distribution (∼100 nm), high drug loading (80 %), and excellent ability to target liver cancer cells. In vitro experiments demonstrated that Cel@Lip-Arg considerably suppressed the activity of HuH7 (hepatoma) cells but had a negligible effect on L02 (normal) cells. Cel@Lip-Arg induced ferroptosis in hepatoma cells by promoting transferrin receptor expression, inhibiting system xc- and glutathione peroxidase 4, and favoring intracellular peroxide accumulation. In vivo experiments revealed that Cel@Lip-Arg plays a therapeutic role by inducing ferroptosis. Compared to Cel, Cel@Lip-Arg had a higher anti-hepatoma activity and effectively reduced the toxicity of Cel in mice. Cel@Lip-Arg-induced ferroptosis was concluded to be an attractive strategy for the precise treatment of HCC.
Collapse
Affiliation(s)
- Manman Qi
- School of Medicine, Shanghai University, Shanghai 200444, PR China
| | - Kai Zhang
- School of Medicine, Shanghai University, Shanghai 200444, PR China
| | - Xue Zhang
- School of Basic Medicine, Ningxia Medical University, Ningxia 750004, PR China
| | - Yuzhao Zhu
- Shanghai Universal Medical Imaging Diagnostic Center, Shanghai University, Shanghai 200233, PR China
| | - Banglan Cai
- School of Basic Medicine, Ningxia Medical University, Ningxia 750004, PR China
| | - Chao Wang
- Shanghai Universal Medical Imaging Diagnostic Center, Shanghai University, Shanghai 200233, PR China
| | - Gang Zhao
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Denghai Zhang
- School of Medicine, Shanghai University, Shanghai 200444, PR China.
| | - Jian Zhang
- School of Medicine, Shanghai University, Shanghai 200444, PR China; Shanghai Universal Medical Imaging Diagnostic Center, Shanghai University, Shanghai 200233, PR China.
| |
Collapse
|
89
|
Wang S, Yang C, Zhang Y, Hu Y, Xiao L, Ding W, Qiu B, Li F. Co-encapsulated Ce6 + CpG and biopeptide-modified liposomes for enhanced transdermal photo-immunotherapy of superficial tumors. Mater Today Bio 2025; 32:101669. [PMID: 40177379 PMCID: PMC11964550 DOI: 10.1016/j.mtbio.2025.101669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 04/05/2025] Open
Abstract
Cancer immunotherapy encounters challenges of a low treatment response rate due to the tumor immunosuppressive microenvironment and immune-related adverse events caused by off-target immunotherapy agents delivered through systemic administration in clinical practice. Photodynamic therapy (PDT) offers a viable approach to improve the immunotherapy efficacy through inducing immunogenic tumor cell death and is particularly advantageous in superficial tumor treatment. Therefore, leveraging integrated nanomaterials for photo-immunotherapy appears to be an ideal strategy to improve therapeutic outcome. In this study, we develop a transdermal-enhancing peptide (TD)-modified cationic liposome that simultaneously encapsulated with photosensitizer chlorine 6 (Ce6) and immunoadjuvant CpG, denoted as Ce6/CpG@Lip-TD, to mediate photo-immunotherapy of superficial tumors via the skin. The functionalization of TD peptide and positively charged surface endow the liposomes enhanced skin penetration capability. The combination of Ce6 and CpG within the liposomes synergistically potentiates the photo-immunotherapy effect when exposed to laser irradiation. In both melanoma and breast cancer murine models, Ce6/CpG@Lip-TD demonstrated substantial tumor-suppressing properties, along with an augmented systemic immune response against distal tumors. As a topical therapeutic agent, Ce6/CpG@Lip-TD circumvents the regulatory challenges associated with the systemic delivery of nanomaterials and significantly reduces systemic side effects, holding great promise for rapid translation into clinical applications.
Collapse
Affiliation(s)
- Shaozhen Wang
- Medical Imaging Center, Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Chen Yang
- Medical Imaging Center, Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yuanyuan Zhang
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
| | - Yi Hu
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230027, China
| | - Lan Xiao
- Department of Gynecology Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
| | - Weiping Ding
- Medical Imaging Center, Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Bensheng Qiu
- Medical Imaging Center, Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Fenfen Li
- Medical Imaging Center, Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
90
|
Zhang Z, Xiong R, Hu Q, Zhang Q, Wang S, Chen Y. Review on anti-tumour lipid nano drug delivery systems of traditional Chinese medicine. J Drug Target 2025; 33:704-716. [PMID: 39743936 DOI: 10.1080/1061186x.2024.2448708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/11/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
In recent years, the use of traditional Chinese medicine (TCM) in the treatment of cancer has received widespread attention. Treatment of tumours using TCM can effectively reduce the side effects of anti-tumour drugs, meanwhile to improve the treatment efficacy of patients. However, most of the active ingredients in TCM, such as saponins, alkaloids, flavonoids, volatile oils, etc., have defects such as low bioavailability and poor solubility in clinical application, which seriously restrict the application of TCM. Meanwhile, the encapsulation of TCM into lipid nano-delivery systems for cancer therapy has received much attention. Lipid nano-delivery systems are obtained by using phospholipids as the base material and adding other auxiliary materials under a certain preparation process, including, for example, liposomes, solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), microemulsions, and self-microemulsion drug delivery systems (SMEDDS), can resolve the application problems of TCM by improving the efficacy of active ingredients of TCM and reducing the toxicity of anti-tumour drugs. This paper focuses on the categories, development status, and research progress of lipid nano delivery system of TCM, aiming to provide a certain theoretical basis for further in-depth research and rational application of these systems.
Collapse
Affiliation(s)
- Ziwei Zhang
- School of Pharmacy, Wannan Medical College, Wuhu, China
- Institute of Synthesis and Application of Medical Materials, Wannan Medical College, Wuhu, China
| | - Rui Xiong
- School of Pharmacy, Wannan Medical College, Wuhu, China
- Institute of Synthesis and Application of Medical Materials, Wannan Medical College, Wuhu, China
| | - Qiyan Hu
- School of Pharmacy, Wannan Medical College, Wuhu, China
- Institute of Synthesis and Application of Medical Materials, Wannan Medical College, Wuhu, China
| | - Qiang Zhang
- School of Pharmacy, Wannan Medical College, Wuhu, China
- Institute of Synthesis and Application of Medical Materials, Wannan Medical College, Wuhu, China
| | - Shaozhen Wang
- School of Pharmacy, Wannan Medical College, Wuhu, China
- Institute of Synthesis and Application of Medical Materials, Wannan Medical College, Wuhu, China
| | - Yunyan Chen
- School of Pharmacy, Wannan Medical College, Wuhu, China
- Institute of Synthesis and Application of Medical Materials, Wannan Medical College, Wuhu, China
| |
Collapse
|
91
|
Zhao L, Wang M, Sun Y, Xu J, Fu Q, Xiao W. pH-responsive nanovesicles capable of remodeling the tumor microenvironment enable activatable near-infrared-II fluorescence image-guided enhanced radiotherapy. Mater Today Bio 2025; 32:101725. [PMID: 40255584 PMCID: PMC12008130 DOI: 10.1016/j.mtbio.2025.101725] [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: 02/12/2025] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/22/2025] Open
Abstract
Traditional radiotherapy (RT) lacks the precision to distinguish between tumor and normal tissues, leading to inevitable X-ray-induced side effects in patients. Therefore, it is crucial to develop integrated imaging and therapeutic modalities that can reduce side effects on surrounding healthy tissues while enhancing susceptibility to tumor tissues. In this study, we developed a pH-responsive nanodrug (AuNRs-Mn3O4-Ag2S Ve) by self-assembling the second near-infrared (NIR-II, 950-1700 nm) fluorescent probe Ag2S quantum dots (QDs), multifunctional nanozyme Mn3O4 nanoparticles (NPs), and radiosensitizer gold nanorods (AuNRs) into a single nanoplatform via an emulsion process. This nanodrug enables precise tumor localization for accurately guided RT and multi-angle sensitization of RT. Upon intravenous administration, the nanodrug disintegrates in the tumor area due to the pH-sensitive polymer P4VP, releasing Ag2S QDs which are specifically activated by the acidic environment, thereby "turning on" the NIR-II fluorescence signal. The optimal timing of the NIR-II fluorescence signal within the tumor region after intravenous injection was investigated, providing a reference for guided RT. In vitro and in vivo experiments confirmed the efficient enhancement of tumor radiosensitization by AuNRs and Mn3O4 NPs. The specific imaging modality that transitions the fluorescence signal from "off" to "on" has been successfully implemented, addressing the limitations of conventional RT and enhancing radiosensitivity. The integration of imaging and therapeutic approaches in this study presents a promising modality for image-guided tumor RT.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
- Institute of Chronic Disease, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Mengzhen Wang
- Institute of Chronic Disease, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yang Sun
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Jinpeng Xu
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Qinrui Fu
- Institute of Chronic Disease, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Wenjing Xiao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
92
|
Duan H, Li Y, Zheng X, Hou J, Tao H, Liu X, Dai M, He S. Paeonol enhances a recombinant EGFR-targeted fusion protein-drug conjugate induced antitumor efficacy in esophageal cancer. Biochem Pharmacol 2025; 236:116856. [PMID: 40054783 DOI: 10.1016/j.bcp.2025.116856] [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: 12/02/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Esophageal cancer (EC) is a gastrointestinal cancer with high morbidity and mortality, along with a low 5-year survival rate, which urgently requires the discovery of new drugs for prevention and treatment. Our previous studies have found a novel EGFR-targeted fusion protein-drug conjugate, Fv-LDP-D3-AE, which exhibits significant inhibitory activity against esophageal cancer. However, the effectiveness of monotherapy still faces major challenges in clinical translation for esophageal cancer treatment. Therefore, there is an urgent need to identify a candidate anti-tumor drug that can be combined with Fv-LDP-D3-AE to enhance therapeutic efficacy. In this study, we report a novel combination treatment regimen of paeonol with Fv-LDP-D3-AE, using human esophageal cancer cells KYSE70 and EC109 for in vitro studies and establishing a BALB/c nude mouse xenograft model for in vivo experiments to investigate the anti-tumor efficacy and potential mechanisms of the combination therapy in esophageal cancer. The results indicated that the combined treatment emerged a synergistic effect, which could effectively inhibit the proliferation, migration, and invasion of esophageal cancer cells, induce more obvious cell apoptosis and DNA damage, and suppress tumor growth in the xenograft mouse model with a tumor inhibition rate of 76%. This may be attributed to the combination therapy simultaneously inhibiting the EGFR/AKT/mTOR signaling pathway and downregulating the expression of nucleolin. Overall, these findings suggest that paeonol could synergize with Fv-LDP-D3-AE to enhance anti-esophageal cancer efficacy, which may be a promising therapeutic strategy for esophageal cancer.
Collapse
Affiliation(s)
- Huaiyu Duan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yuting Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Xue Zheng
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Junqi Hou
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Hongyu Tao
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, China
| | - Xiujun Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Min Dai
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| | - Shiming He
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
93
|
Guo Z, He L, Wang W, Tian S, Lin R. FUT2-dependent fucosylation of LAMP1 promotes the apoptosis of colorectal cancer cells by regulating the autophagy-lysosomal pathway. Cancer Lett 2025; 619:217643. [PMID: 40112906 DOI: 10.1016/j.canlet.2025.217643] [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: 12/23/2024] [Revised: 03/06/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Fucosyltransferase 2 (FUT2) is an enzyme that adds fucose to proteins or lipids via α-1,2-fucosylation in the intestinal mucosa. While FUT2 deficiency is linked to increased susceptibility to inflammatory bowel disease (IBD), its role in colorectal cancer (CRC) is unclear, and the molecular mechanisms involved remain largely unknown. We established an azoxymethane (AOM) and dextran sulfate sodium (DSS) model to induce CRC. FUT2 expression was assessed in human CRC tissues, AOM/DSS-induced mouse models, and CRC cell lines using qRT-PCR, western blotting, and UEA-I staining. FUT2 knockout (FUT2△IEC) mice were treated with AOM/DSS, and FUT2-overexpressing CRC cells were created to evaluate the effects of FUT2 on apoptosis in both in vitro and in vivo settings through Western blot analyses and functional assays. N-glycoproteomics, UEA-I chromatography, and co-immunoprecipitation were utilized to identify regulatory mechanisms and target fucosylated proteins. FUT2 expression and α-1,2-fucosylation were significantly decreased in CRC. FUT2 deficiency worsened AOM/DSS-induced CRC and reduced tumor apoptosis, while FUT2 overexpression induced apoptosis and inhibited proliferation in CRC cells and xenografts. Mechanistically, FUT2 appears to suppress autophagy by impairing lysosomal function and directly targeting and fucosylating LAMP1, contributing to lysosomal dysfunction. Our study reveals a fucosylation-dependent antitumor mechanism of FUT2 in CRC, suggesting potential therapeutic strategies for CRC treatment.
Collapse
Affiliation(s)
- Zijun Guo
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingnan He
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New Area, Shanghai, China
| | - Weijun Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuxin Tian
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
94
|
Reddy R, Gaiwak V, Goda JS, Teni T. 'Nelfinavir sensitizes a clinically relevant chemo-radioresistant cervical cancer in-vitro model by targeting the AKT-USP15/USP11-HPV16 E6/E7 axis. Biochem Biophys Rep 2025; 42:101987. [PMID: 40242402 PMCID: PMC12002845 DOI: 10.1016/j.bbrep.2025.101987] [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: 10/29/2024] [Revised: 03/12/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Resistance to standard therapies is a major challenge in managing cervical cancer, often leading to systemic relapse. This study aimed to develop an in-vitro model of chemo-radioresistant cervical cancer that mimics clinical conditions and also explore the therapeutic potential of the repurposed drug nelfinavir, an HIV protease inhibitor. HPV16-positive SiHa cervical cancer cells were subjected to concurrent cisplatin and ionizing radiation, to simulate the clinical treatment regimen for locally advanced cervical cancer. The resulting chemo-radioresistant subline exhibited increased IC50-value, D0 dose, and a higher Resistance Index compared to parent cells, indicating resistance development. Notably, elevated HPV16 E6/E7 expression in resistant sublines suggested a role for HPV16 in resistance acquisition. Treatment with nelfinavir significantly reduced the IC50-value and D0 dose in resistant cells. Additionally, exposure to nelfinavir or AKT inhibitor IV showed significant decrease in AKT, USP15, USP11 and HPV16 E6/E7 proteins. Furthermore, siRNA mediated knockdown of USP15 and USP11 in resistant cells resulted in significant reduction of HPV16 E6 and E7 oncoproteins respectively. Thus, mechanistically nelfinavir sensitized resistant cervical cancer cells by inhibiting the AKT-USP15/USP11-HPV16 E6/E7 pathway. Overall, this study successfully established a chemo-radioresistant SiHa cell model, providing a platform for investigating resistance mechanisms. It also highlights nelfinavir's potential as a therapeutic agent in overcoming chemo-radioresistance in cervical cancer.
Collapse
Affiliation(s)
- Reshma Reddy
- Teni Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, 400094, India
| | - Vagmi Gaiwak
- Teni Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
| | - Jayant Sastri Goda
- Department of Radiation Oncology, Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, 400094, India
| | - Tanuja Teni
- Teni Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, 400094, India
| |
Collapse
|
95
|
Le HDM, Vo DT, Do HT, Le HNG, Phan CC, Nguyen DT, Le QND. Hepatectomy in a young patient with advanced hepatocellular carcinoma and poor prognostic imaging features: A case of recurrence-free survival. Radiol Case Rep 2025; 20:2704-2709. [PMID: 40151292 PMCID: PMC11937630 DOI: 10.1016/j.radcr.2025.02.085] [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: 11/15/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
A 45-year-old male with chronic hepatitis B presented with an advanced hepatocellular carcinoma (HCC) occupying the entire left liver and invading the left portal vein. Despite multiple poor prognostic imaging features, including vascular invasion, corona enhancement, an incomplete capsule, intratumoral necrosis, intratumoral arteries, and irregular tumor borders, the patient elected to undergo a left hepatectomy. Although Barcelona Clinic Liver Cancer (BCLC) staging classified the case as stage C, a resection was successfully performed. Remarkably, 6 years postsurgery, the patient remains recurrence-free. This report highlights a rare, fortunate outcome in a high-risk HCC case and underscores the potential of surgical intervention even in advanced HCC.
Collapse
Affiliation(s)
- Huyen Duy Mai Le
- Department of Diagnostic Imaging, University Medical Center, Ho Chi Minh City, Vietnam
| | - Duc Tan Vo
- Department of Diagnostic Imaging, University Medical Center, Ho Chi Minh City, Vietnam
- Department of Diagnostic Imaging, University of Medicine and Pharmacy, Ho Chi Minh City, Vietnam
| | - Hai Trong Do
- Department of General Surgery, University of Medicine and Pharmacy, Ho Chi Minh City, Vietnam
| | - Hy Nguyen Gia Le
- Department of Diagnostic Imaging, University Medical Center, Ho Chi Minh City, Vietnam
| | - Chien Cong Phan
- Department of Diagnostic Imaging, University Medical Center, Ho Chi Minh City, Vietnam
| | - Duy Thanh Nguyen
- Department of Diagnostic Imaging, University Medical Center, Ho Chi Minh City, Vietnam
| | - Quynh Nguyen Diem Le
- Department of Diagnostic Imaging, University Medical Center, Ho Chi Minh City, Vietnam
| |
Collapse
|
96
|
Bokhary RY. Prevalence of HER2 expression and its association with clinicopathological parameters in gastric and gastroesophageal junction adenocarcinoma: A 10?year experience of an academic center. Mol Clin Oncol 2025; 22:49. [PMID: 40242368 PMCID: PMC12001012 DOI: 10.3892/mco.2025.2844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
HER2 overexpression is a marker for targeted therapy in adenocarcinoma of the gastroesophageal junction (GEJ) and stomach. The present study aimed to evaluate the frequency of HER2 overexpression with reference to clinicopathological characteristics in subjects from King Abdulaziz University Hospital, Jeddah, Saudi Arabia over a 10-year period. A retrospective cross-sectional study was conducted on all biopsy and resection specimens diagnosed with either gastric cancer (GC) or GEJ adenocarcinomas from patients between January 2014 and December 2023 that had a final pathology report. Demographic characteristics of 122 patients, including age and sex, were collected, along with pathological details such as tumor grade, histological subtype and HER2 status. χ2 test was used to analyze the association between collected clinicopathological characteristics and HER2 status of the tumor. Most patients were aged 40-60 years. Males constituted 66% of the patients, and the ethnic distribution between Saudi and non-Saudi was almost equal. The most common subtype of cancer was the intestinal type (49%), and the majority of cases were poorly differentiated (64%). HER2 status was assessed in only 61% of cases, with 13.5% showing gene amplification. There was no significant association found between HER2 status and clinicopathological features.
Collapse
Affiliation(s)
- Rana Y. Bokhary
- Department of Pathology, Faculty of Medicine, King Abdulaziz University & King Abdulaziz University Hospital, Jeddah 21589, Saudi Arabia
| |
Collapse
|
97
|
Lin T, Li Z, Yuan J, Ren T, Pang W, Xu S. Design, synthesis and evaluation of diphenyl ether-based kaiso inhibitors with enhanced potency. Bioorg Med Chem Lett 2025; 121:130158. [PMID: 40049243 DOI: 10.1016/j.bmcl.2025.130158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/10/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025]
Abstract
Kaiso, a potential target for the treatment of lung cancer. Our research focuses on Kaiso inhibitros. Through virtual screening and molecular dynamic simulations, we discovered a promising Kaiso inhibitor called compound 5 (ZINC20577650). By modifying the structure of compound 5, a series of novel Kaiso inhibitors that contain a diphenyl ether ring were synthesized. Among them, compound 20 exhibited the strongest inhibitory activity against A549 cells (IC50 = 0.34 μM). Notably, its inhibitory activity surpassed that of the positive control MIRA-1 (IC50 = 654.065 μM). Molecular docking and dynamic studies revealed that the binding of the compound's amino and ester moieties to the active site of kaiso protein, as well as the extension of the benzene ring towards the Asn561 position in the cavity, contributed significantly to its potency. These findings provide valuable insights for the development of new Kaiso inhibitors.
Collapse
Affiliation(s)
- Taofeng Lin
- College of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Zhongqi Li
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University. Nanchang, China
| | - Juanchan Yuan
- College of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Tinfeng Ren
- College of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Wan Pang
- College of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China.
| | - Songhui Xu
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University. Nanchang, China; Key Laboratory of Urinary System Diseases of Jiangxi Province, Nanchang, China.
| |
Collapse
|
98
|
Xu H, Fu X, Wang S, Ge Y, Zhang L, Li J, Zhang F, Yang Y, He Y, Sun Y, Gao A. Immunoglobulin-like transcript 5 polarizes M2-like tumor-associated macrophages for immunosuppression in non-small cell lung cancer. Int J Cancer 2025; 156:2225-2236. [PMID: 39910654 PMCID: PMC11970544 DOI: 10.1002/ijc.35360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 11/28/2024] [Accepted: 12/04/2024] [Indexed: 02/07/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have shifted the treatment paradigm of non-small cell lung cancer (NSCLC) over the last decade. Despite notable therapeutic advancements in responders, the response rate remains limited owing to the immunosuppressive tumor microenvironment (TME). Therefore, to improve the efficacy of ICIs, it is essential to explore alternative targets or signals that mediate immunosuppression. Immunoglobulin-like transcript (ILT) 5 is a negative regulator of immune activation in myeloid cells. However, the expression and function of ILT5 in NSCLC remain unknown. Here, we found that ILT5 was highly expressed in tumor-associated macrophages (TAMs) of NSCLC tissues and predicted poor patient survival. Functionally, ILT5 induces the M2-like polarization of TAMs, which subsequently decreases the density of T cells, and increases FOXP3+T cell accumulation, leading to an immunosuppressive TME. The combination of ILT5 expression with M2-like TAM density is a more reliable biomarker of patient survival than ILT5 expression alone. ILT5 knockout mitigates the reprogramming of TAM and T cell subsets toward immunosuppressive phenotypes and inhibits tumor growth in vivo. These findings highlight that ILT5 is a potential immunotherapeutic target and a promising prognostic biomarker for NSCLC.
Collapse
Affiliation(s)
- Huijun Xu
- Jinan Central HospitalShandong UniversityJinanShandongChina
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Xuebing Fu
- Department of Thoracic Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Shuyun Wang
- Phase I Clinical Research Center, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Yihui Ge
- Phase I Clinical Research Center, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Lu Zhang
- Department of OncologyThe Fourth People's Hospital of ZiboZiboShandongChina
| | - Juan Li
- Phase I Clinical Research Center, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Fang Zhang
- Department of OncologyCentral Hospital affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Yang Yang
- Department of Ultrasound, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Yifu He
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Yuping Sun
- Phase I Clinical Research Center, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Aiqin Gao
- Department of Thoracic Radiation Oncology, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| |
Collapse
|
99
|
Heirman AN, Duimel SLL, Tellman RS, Dirven R, Linn AJ, Halmos GB, van Son RJJH, van der Molen L, Balm AJM, van Weert JCM, van den Brekel MWM. Exploring the state of shared decision-making in head and neck oncology: Assessing treatment communication. PATIENT EDUCATION AND COUNSELING 2025; 135:108733. [PMID: 40088519 DOI: 10.1016/j.pec.2025.108733] [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: 05/29/2024] [Revised: 02/26/2025] [Accepted: 03/02/2025] [Indexed: 03/17/2025]
Abstract
OBJECTIVE To explore the state of shared decision-making (SDM) in head and neck (HN) oncology by investigating the extent to which SDM is currently employed by HN surgeons and how the perceived levels of SDM relate to the observed levels of SDM. Additionally, surgeon and patient perspectives on patient involvement in SDM and potential associations with observed levels of SDM were explored. METHODS Perceived level of SDM and patient involvement were measured by SDM-Q-9/SDM-Q-Doc resp. Control Preference Scale. Observed SDM was measured by analyzing audiotaped consultations (N = 42) using the OPTIONmcc+ instrument. Univariate linear analyses were conducted to identify possible associations with surgeon-observed SDM. RESULTS Median perceived SDM scores of surgeons (74.4 %) and patients (71.1 %) were relatively high, whereas observed median OPTION-scores were moderate (surgeons 48 %, patients 42 %, caregivers 24 %). Consultation time and patient OPTION-score were positively associated with surgeon OPTION-score. CONCLUSION Surgeons and patients seem to overestimate the extent of SDM compared to the observed reality. Patients' goals, values and preferences need to be addressed more during consultations. PRACTICE IMPLICATIONS The findings can be used to raise awareness of SDM among surgeons to improve their skills. Routine training in medical education can benefit from effective integration of SDM principles during consultations.
Collapse
Affiliation(s)
- Anne N Heirman
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Song L L Duimel
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Amsterdam School of Communication Research/ASCoR, Department of Communication Science, University of Amsterdam, Amsterdam, the Netherlands.
| | - Roosmarijn S Tellman
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Richard Dirven
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Annemiek J Linn
- Amsterdam School of Communication Research/ASCoR, Department of Communication Science, University of Amsterdam, Amsterdam, the Netherlands.
| | - Gyorgy B Halmos
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Rob J J H van Son
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Amsterdam Center of Language and Communication, University of Amsterdam, Amsterdam, the Netherlands.
| | - Lisette van der Molen
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Alfons J M Balm
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Oral and Maxillofacial Surgery, Amsterdam University Medical Center, Amsterdam, the Netherlands.
| | - Julia C M van Weert
- Amsterdam School of Communication Research/ASCoR, Department of Communication Science, University of Amsterdam, Amsterdam, the Netherlands.
| | - Michiel W M van den Brekel
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Amsterdam Center of Language and Communication, University of Amsterdam, Amsterdam, the Netherlands; Department of Oral and Maxillofacial Surgery, Amsterdam University Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
100
|
Chen X, Hong F, Shen Y, Xia H, Shi L, Jiang Z, Xu RA. Inhibitory effects of nimodipine, nitrendipine and felodipine on tamoxifen metabolism and molecular docking. Biochem Pharmacol 2025; 236:116854. [PMID: 40054784 DOI: 10.1016/j.bcp.2025.116854] [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: 07/19/2024] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Tamoxifen, a selective estrogen receptor modulator (SERM) used in breast cancer therapy, requires metabolic activation by CYP3A4 to exert its biological effects. This study evaluated the effects of calcium channel blockers nimodipine, nitrendipine and felodipine on tamoxifen metabolism by studying their interactions with tamoxifen in vitro and in vivo. Rat liver microsomes (RLM) and human liver microsomes (HLM) were used in this study to evaluate the inhibitory potential of nimodipine, nitrendipine and felodipine on tamoxifen metabolism in vitro. A total of 28 cardiovascular drugs, including calcium channel blockers, were screened in an RLM incubation system in vitro. In RLM, nimodipine, nitrendipine and felodipine had half-maximum inhibitory concentration (IC50) values of 5.55 µM, 11.86 µM and 7.71 µM, respectively. In HLM, the IC50 values were increased to 20.38 µM, 30.06 µM, and 44.45 µM for nimodipine, nitrendipine and felodipine, respectively. The kinetic assays indicated that nimodipine and felodipine inhibited the metabolism of tamoxifen in a competitive way, whereas nitrendipine showed non-competitive inhibition in RLM. However, felodipine exhibited non-competitive inhibition, and nimodipine and nitrendipine showed competitive inhibition in HLM. Pharmacokinetic studies in rats revealed that pretreatment with nimodipine and nitrendipine significantly increased the systemic exposure of tamoxifen, as demonstrated by increasing the area under the curve (AUC), the maximum concentration (Cmax) and decreasing the clearance (CLz/F). Finally, molecular docking studies supported these findings, showing potential interactions at the active site of CYP3A4. These results suggested the necessity for careful monitoring and possible dose adjustments of tamoxifen when co-administered with calcium channel blockers in clinic.
Collapse
Affiliation(s)
- Xiaohai Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fengsheng Hong
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuxin Shen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hailun Xia
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lu Shi
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zheli Jiang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ren-Ai Xu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|