Medical OncologyPub Date : 2025-08-13DOI: 10.1007/s12032-025-02918-8
Lei Yang, Jinhai Tang, Yuteng Chi, Huiming Li, Meiyue Zhang, Haixin Zhang, Xiaoxuan Ma, Jun Cheng, Chao Gao
{"title":"Neoadjuvant chemoradiotherapy combined with immunotherapy: a promising strategy for MSS/pMMR locally advanced rectal cancer.","authors":"Lei Yang, Jinhai Tang, Yuteng Chi, Huiming Li, Meiyue Zhang, Haixin Zhang, Xiaoxuan Ma, Jun Cheng, Chao Gao","doi":"10.1007/s12032-025-02918-8","DOIUrl":"10.1007/s12032-025-02918-8","url":null,"abstract":"<p><p>Research prospects of neoadjuvant immunotherapy based on different microsatellite status in locally advanced rectal cancer patients are a key area of current research. Neoadjuvant chemoradiotherapy followed by total mesorectal excision is the first-line treatment for locally advanced rectal cancer (LARC). In recent years, extensive clinical studies have demonstrated the significant importance of immunotherapy in improving survival rates for LARC patients with microsatellite instability (MSI), while also highlighting the potential benefits for those with microsatellite stability (MSS). Microsatellite status is one of the key markers for the classification of LARC subtypes and is also the decisive basis for the formulation of immunotherapy strategies. Although the important therapeutic value of immunotherapy in LARC patients with MSI has been strongly confirmed, LARC patients with MSS do not appear to benefit from immune monotherapy. The current article seeks to synthesize the progress of immunotherapy for LARC based on MSS. By highlighting these comparative insights, we aim to advance our understanding of how immunotherapy may address the unique needs of LARC patients with MSS profile characteristics. Furthermore, this research paves the way for future studies that can guide clinical decision-making in treating LARC patients with MSS.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 10","pages":"428"},"PeriodicalIF":3.5,"publicationDate":"2025-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144835563","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-08-12DOI: 10.1007/s12032-025-02978-w
Şeyma Taştemur, Ahmet Ozan Kaleci, Ayşegül Öztürk, Ali Sefa Mendil
{"title":"Astaxanthin promotes apoptosis by suppressing growth signaling pathways in HT-29 colorectal cancer cells.","authors":"Şeyma Taştemur, Ahmet Ozan Kaleci, Ayşegül Öztürk, Ali Sefa Mendil","doi":"10.1007/s12032-025-02978-w","DOIUrl":"10.1007/s12032-025-02978-w","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is the third most frequently diagnosed malignancy globally and ranks second in cancer-related mortality. Despite advancements in therapeutic approaches, the need for novel, effective and less toxic treatment strategies remains critical. Astaxanthin (ATX), a naturally occurring xanthophyll carotenoid, has attracted attention due to its strong antioxidant, anti-inflammatory and anti-cancer properties. This study aimed to evaluate the antiproliferative and pro-apoptotic effects of ATX on CRC through its influence on key molecular pathways, involved in tumorigenesis. The human colorectal adenocarcinoma cell line HT-29 was treated with varying concentrations of ATX (10 µM and 20 µM) for 24 h. Cell viability was assessed using the XTT assay. The expression levels of HER2, EGFR, ERK1, ERK2 and mTOR were quantified via enzyme-linked immunosorbent assay (ELISA). Immunofluorescence staining was used to evaluate the expression of EGFR and caspase-3 proteins. ATX exhibited significant antiproliferative and pro-apoptotic effects on HT-29 cells, with an IC50 value of 10.98 µM at 24 h. Treatment with ATX (10.98 µM) led to a marked increase in caspase-3 expression and a significant reduction in EGFR levels. Additionally, HER2, ERK1 and ERK2 levels were significantly downregulated, while mTOR expression remained unaffected. Flow cytometry analysis revealed a significant increase in apoptotic cell populations following ATX treatment, compared to the control group. ATX exerts notable antiproliferative and pro-apoptotic effects on CRC cells, potentially through modulation of the EGFR/HER2/ERK signaling pathway. These findings suggest that ATX may serve as a promising candidate for further investigation as an adjunctive or standalone therapeutic agent in the treatment of CRC.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 9","pages":"426"},"PeriodicalIF":3.5,"publicationDate":"2025-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144821996","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-08-12DOI: 10.1007/s12032-025-02971-3
Ru He, Heng Xu, Hao Chi
{"title":"Letter to the editor: comment on \"Integration of immunotherapy and radiotherapy in a therapeutic algorithm for locally advanced squamous cell skin cancer\".","authors":"Ru He, Heng Xu, Hao Chi","doi":"10.1007/s12032-025-02971-3","DOIUrl":"10.1007/s12032-025-02971-3","url":null,"abstract":"","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 9","pages":"427"},"PeriodicalIF":3.5,"publicationDate":"2025-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144821997","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-08-11DOI: 10.1007/s12032-025-02982-0
Zirui Jiang, Joshua Low, Colin Huang, Yong Yue, Christopher Njeh, Oluwaseyi Oderinde
{"title":"18F-FDG PET/CT-based deep radiomic models for enhancing chemotherapy response prediction in breast cancer.","authors":"Zirui Jiang, Joshua Low, Colin Huang, Yong Yue, Christopher Njeh, Oluwaseyi Oderinde","doi":"10.1007/s12032-025-02982-0","DOIUrl":"10.1007/s12032-025-02982-0","url":null,"abstract":"<p><p>Enhancing the accuracy of tumor response predictions enables the development of tailored therapeutic strategies for patients with breast cancer. In this study, we developed deep radiomic models to enhance the prediction of chemotherapy response after the first treatment cycle. 18F-Fludeoxyglucose PET/CT imaging data and clinical record from 60 breast cancer patients were retrospectively obtained from the Cancer Imaging Archive. PET/CT scans were conducted at three distinct stages of treatment; prior to the initiation of chemotherapy (T1), following the first cycle of chemotherapy (T2), and after the full chemotherapy regimen (T3). The patient's primary gross tumor volume (GTV) was delineated on PET images using a 40% threshold of the maximum standardized uptake value (SUVmax). Radiomic features were extracted from the GTV based on the PET/CT images. In addition, a squeeze-and-excitation network (SENet) deep learning model was employed to generate additional features from the PET/CT images for combined analysis. A XGBoost machine learning model was developed and compared with the conventional machine learning algorithm [random forest (RF), logistic regression (LR) and support vector machine (SVM)]. The performance of each model was assessed using receiver operating characteristics area under the curve (ROC AUC) analysis, and prediction accuracy in a validation cohort. Model performance was evaluated through fivefold cross-validation on the entire cohort, with data splits stratified by treatment response categories to ensure balanced representation. The AUC values for the machine learning models using only radiomic features were 0.85(XGBoost), 0.76 (RF), 0.80 (LR), and 0.59 (SVM), with XGBoost showing the best performance. After incorporating additional deep learning-derived features from SENet, the AUC values increased to 0.92, 0.88, 0.90, and 0.61, respectively, demonstrating significant improvements in predictive accuracy. Predictions were based on pre-treatment (T1) and post-first-cycle (T2) imaging data, enabling early assessment of chemotherapy response after the initial treatment cycle. Integrating deep learning-derived features significantly enhanced the performance of predictive models for chemotherapy response in breast cancer patients. This study demonstrated the superior predictive capability of the XGBoost model, emphasizing its potential to optimize personalized therapeutic strategies by accurately identifying patients unlikely to respond to chemotherapy after the first treatment cycle.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 9","pages":"425"},"PeriodicalIF":3.5,"publicationDate":"2025-08-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12339616/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144821995","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-08-11DOI: 10.1007/s12032-025-02986-w
Dilpreet Singh, Satvir Singh, Nitin Tandon
{"title":"Multi-epitope ligand-conjugated nanoparticles for tumor neoantigen targeting: advancing molecular precision in cancer immunotherapy.","authors":"Dilpreet Singh, Satvir Singh, Nitin Tandon","doi":"10.1007/s12032-025-02986-w","DOIUrl":"10.1007/s12032-025-02986-w","url":null,"abstract":"<p><p>Tumor neoantigens, derived from somatic mutations unique to cancer cells, represent a novel class of highly specific targets for precision immunotherapy. Their absence in normal tissues minimizes the risk of central tolerance, offering the potential to elicit tumor-specific immune responses. MEL-NPs are engineered to display multiple neoepitope peptides on their surface as antigenic cargo while incorporating targeting ligands-such as antibodies or aptamers-that promote specific uptake by dendritic cells. This dual-functionalization enables both antigen presentation and active delivery to immune-priming sites. However, their clinical application is often hindered by low abundance, inefficient antigen presentation, and limited delivery to antigen-presenting cells (APC) Nanoparticle-based delivery platforms have emerged as transformative tools to address these challenges by enhancing neoantigen stability, promoting tumor-site accumulation, and improving immune co-stimulation. Among them, multi-epitope ligand-conjugated nanoparticles (MEL-NPs) represent a next-generation strategy that enables modular co-display of multiple neoepitopes and targeted delivery to tumor-infiltrating dendritic cells (DCs). This multivalent configuration enhances antigen uptake, cross-presentation, and activation of polyclonal CD8⁺ and CD4⁺ T cell responses. The review discusses the molecular landscape of neoantigens, advances in nanoparticle engineering, immune activation pathways, and preclinical/clinical data supporting MEL-NPs. By integrating molecular specificity with immunological breadth, MEL-NPs offer a promising platform to overcome tumor heterogeneity and immune evasion in personalized cancer immunotherapy.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 9","pages":"424"},"PeriodicalIF":3.5,"publicationDate":"2025-08-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144817082","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-08-09DOI: 10.1007/s12032-025-02973-1
Jitendra Gupta, Bahaa Ibrahim Saeed, Ashok Kumar Bishoyi, Ali G Alkhathami, Shodiyev Asliddin, Deepak Nathiya, M Ravi Kumar, Deepak Bhanot, Amera Bekhatroh Rashed, Yasser Fakri Mustafa
{"title":"From cell cycle control to cancer therapy: exploring the role of CDK1 and CDK2 in tumorigenesis.","authors":"Jitendra Gupta, Bahaa Ibrahim Saeed, Ashok Kumar Bishoyi, Ali G Alkhathami, Shodiyev Asliddin, Deepak Nathiya, M Ravi Kumar, Deepak Bhanot, Amera Bekhatroh Rashed, Yasser Fakri Mustafa","doi":"10.1007/s12032-025-02973-1","DOIUrl":"10.1007/s12032-025-02973-1","url":null,"abstract":"<p><p>The cyclin-dependent kinase (CDK) family comprises one of the most common types of serine/threonine kinases responsible for controlling many cellular processes through the action of the dynamics of these enzymes. The CDKs are regulators of cellular processes, ranging from cell cycle progression, an integral step of cellular lifespan involved in its division and growth, to the expression of genes and the transfer of genetic information from DNA to RNA. CDK-related pathology is primarily based on the dysregulation of CDK, resulting in improper coordination of the cell cycle and leading to uncontrolled proliferation. The CDK1 involved in the G2 to M-phase transition interacts with CENPF, PVT1, and TFCP2L1, which affects chromosome segregation and cell proliferation. Besides the previously described partners, namely PRDX2 and C/EBPα, the CDK2 responsible for the G1/S transition has also been shown to bind TBK1 and modulate DNA synthesis and cell cycle checkpoints. Such mechanisms of action contribute to oncogenesis through interactions, including roles for lncRNAs and miRNAs in modulating the expression and activity of CDKs. Their importance in cancer has made CDK1/2 critical candidates for anticancer drug targeting. The dysregulation of CDK1/2 manifests in cancer pathology, which is, in turn, associated with an uncoordinated normal cell cycle and resultant proliferation. This study aims to provide a comprehensive overview of the diverse functions of CDK1 and CDK2 in cancer, thereby facilitating the exploration of their therapeutic potential and the development of novel cancer therapy designs. The complexity of targeting CDK1 and CDK2 highlights the importance of their regulation during cancer development. It indicates their substantial promise as valid targets for therapy, in vitro and vivo studies, and human sample analysis.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 9","pages":"422"},"PeriodicalIF":3.5,"publicationDate":"2025-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144804424","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-08-09DOI: 10.1007/s12032-025-02974-0
Farah Ijaz, Shaukat Ali, Asim Pervaiz, Muhammad Summer
{"title":"Anticancer efficacy of sericin (silkworm protein) and sericin chitosan conjugated silver nanoparticles against colorectal cancer.","authors":"Farah Ijaz, Shaukat Ali, Asim Pervaiz, Muhammad Summer","doi":"10.1007/s12032-025-02974-0","DOIUrl":"10.1007/s12032-025-02974-0","url":null,"abstract":"<p><p>Sericin is a globular protein known to have antioxidant potential due to presence of amino, carboxyl and hydroxyl groups in its structure. This study was designed to investigate the antiproliferative and apoptotic potential of sericin and sericin chitosan conjugated silver nanoparticles against colorectal cancer cells. To investigate the antiproliferative and apoptotic activity of sericin and sericin chitosan conjugated silver nanoparticles (SChiAgNPs), three human colorectal cancer cell lines (SW480, SW620, HCT116) were used. Sericin was isolated by the degumming process followed by the characterization by using FTIR, UV, XRD, and SEM techniques to confirm the isolation process and successful synthesis of SChiAgNPs. MTT assay was carried out to analyze the antiproliferative activities, while expression profiling of the genes i.e., GADD45A, BCL2, and TNF was assessed by qRT-PCR analysis. Sericin (S-Ext) and SChiAgNPs showed significant antiproliferative activities in SW480, SW620 and HCT 116 cells. Overall, there was 29-34 inhibition of viability for sericin S-Ext and 35-43 for SChiAgNPs in the three cell lines in comparison to untreated control. Expression profiling indicated the significant stimulation of GADD45A, BCL-2 and TNF genes expression in SW480, SW620 and HCT 116 cells. The GADD 45 A showed induction by 1.43-1.71-fold in SW480, 1.09-1.56-fold in SW620 and 1.25-4.55-fold in HCT 116 cells in response to treatment groups. The BCL2 showed the induction by 1.35-2.53, 1.38-3.1, and 2.32-3.76-fold in SW480, SW620, and HCT116 cells, respectively. TNF was induced by a factor of 3.9-6.43, 2.53-5.41, and 2.7-5.31-fold in in SW480, SW620, and HCT116 cells, respectively, after the exposure with compounds. Sericin and S-ChiAgNPs, showed significant growth inhibition and gene expression profiling modifications in the colorectal cancer cells. The findings provide evidence about sericin and its nanoparticle conjugates as potential anticancer medicine for colorectal cancer.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 9","pages":"423"},"PeriodicalIF":3.5,"publicationDate":"2025-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144812185","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-08-09DOI: 10.1007/s12032-025-02979-9
Si-Yu Chen, Si-Ying Chen, Shu Yang, Yi Li, Sheng-Yong Yang
{"title":"ADAR1-mediated RNA editing in breast cancer: molecular mechanisms and therapeutic implications.","authors":"Si-Yu Chen, Si-Ying Chen, Shu Yang, Yi Li, Sheng-Yong Yang","doi":"10.1007/s12032-025-02979-9","DOIUrl":"10.1007/s12032-025-02979-9","url":null,"abstract":"<p><p>Breast cancer (BC) continues to be among the most widespread and clinically demanding cancers globally, resulting in significant illness and death, even with progress in early detection and treatment approaches. Traditional therapeutic modalities, including surgery, chemotherapy, and targeted therapies, often fail in the face of drug resistance, metastasis, and the invasion. Consequently, a pressing demand exists for new biomarkers and treatment approaches to enhance clinical results. A recent area of focus in cancer research is RNA editing, particularly the role of Adenosine deaminase acting on RNA1 (ADAR1) which operates as an RNA editing enzyme that catalyzes adenosine-to-inosine conversion within double-stranded RNA substrates. This process affects gene expression and various cellular functions including RNA stability, splicing, and translation. Recent studies have highlighted the important function of ADAR1 editing in several types of cancer, including breast cancer, by regulating key pathways that are critical to tumor development, proliferation, and therapeutic resistance. This review aims to inspire further research into ADAR1-mediated mechanisms, offering new perspectives to overcome therapeutic resistance and improve clinical outcomes. This review systematically examines the multifaceted roles of ADAR1 in breast cancer progression, with a focus on its upstream regulators and downstream effects on critical pathways. Our analysis not only consolidates current understanding of ADAR1's regulatory networks, but also pointed out the current problems with ADAR1 in breast cancer research. What's more, we also proposed innovative strategies such as ADAR1 inhibitors to advance precision medicine in breast cancer.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 9","pages":"421"},"PeriodicalIF":3.5,"publicationDate":"2025-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144804423","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-08-09DOI: 10.1007/s12032-025-02968-y
Qi Liu, Qian Zhou, Xiaoxuan Yang, Jie Yang, Lizhen Liang, Liang Kang, Yamei Zhang, Yu Deng
{"title":"Tanshinone IIA is synergistic with the PARP inhibitor olaparib in inducing BRCAs-proficient and -deficient triple-negative breast cancer cell apoptosis.","authors":"Qi Liu, Qian Zhou, Xiaoxuan Yang, Jie Yang, Lizhen Liang, Liang Kang, Yamei Zhang, Yu Deng","doi":"10.1007/s12032-025-02968-y","DOIUrl":"10.1007/s12032-025-02968-y","url":null,"abstract":"<p><p>Poly ADP-ribose Polymerase (PARP) inhibitor-based targeted therapy benefits the triple-negative breast cancer (TNBC) patients with Breast cancer susceptibility genes (BRCAs) mutation. However, only about 50% BRCA-mutated TNBC patients respond to PARP inhibitor treatment and 80% TNBC patients are BRCA proficient, which limit clinical application of PARP inhibitor for TNBC treatment. Ataxia-telangiectasia mutated (ATM) is a DNA double-strand break (DSB) sensor to detect and facilitate DSB repair. ATM deficiency sensitizes cancer cells to PARP inhibitor. Currently, none of ATM inhibitor is approved for clinical use largely due to toxicity. Tanshinone IIA (Tan IIA) is a natural compound derived from Salvia miltiorrhiza and has been approved for clinical application for cardiovascular diseases treatment in China. This study aims to evaluate whether Tan IIA could sensitize both BRCAs-proficient and -deficient TNBC cells to PARP inhibitor (olaparib) and explore the underlying molecular mechanisms. We report that Tan IIA synergistically enhances the cytotoxic effects of olaparib in both BRCA-proficient and -deficient TNBC cells. Tan IIA increases DSBs in TNBC cells and subsequently triggers apoptosis by destabilizing ATM. The results suggest that Tan IIA is a potential combinatory drug to enhance PARP inhibitor efficacy in TNBC treatment.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 9","pages":"419"},"PeriodicalIF":3.5,"publicationDate":"2025-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144804427","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-08-09DOI: 10.1007/s12032-025-02965-1
Aohua Zhang, Xiaodi Yin, Jun Ma
{"title":"NK-derived exosomes in anti-tumor strategies.","authors":"Aohua Zhang, Xiaodi Yin, Jun Ma","doi":"10.1007/s12032-025-02965-1","DOIUrl":"10.1007/s12032-025-02965-1","url":null,"abstract":"<p><p>Tumors remain one of the major challenges confronting researchers today. The development of tumor is driven by genetic factors, environmental exposures, and homeostatic disruptions, leading to the transformation of normal cells into malignant ones. Natural killer (NK) cells, a crucial class of immune cells in immunotherapy, can directly recognize and eliminate tumor cells without prior stimulation by tumor antigens. Exosomes derived from NK cells exert anti-tumor effects through multiple mechanisms, including the release of cytotoxic molecules, death receptor ligand-mediated apoptosis, and the secretion of cytokines and other bioactive molecules. However, the therapeutic efficacy of unmodified exosomes is constrained by insufficient active components, suppression by the tumor microenvironment, and a lack of targeting specificity. Drug loading and engineering strategies can enhance their therapeutic potential. In this review, we examine advancements in NK cell-derived exosome (NK-Exos)-based anti-tumor strategies, focusing on studies involving native exosomes, drug-loaded exosomes, and surface-modified exosomes for tumor eradication.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 9","pages":"418"},"PeriodicalIF":3.5,"publicationDate":"2025-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12335398/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144804426","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}