{"title":"A predictive model and mechanistic study of treatment effectiveness in patients newly diagnosed with small cell lung cancer.","authors":"Tianyun Wang, Qiuyang Lu, Diexiao Luo, Chunfang Tao, Jiaqin Liu, Hongbo Zou, Qichao Xie, Rui Kong","doi":"10.3389/fonc.2025.1631490","DOIUrl":"10.3389/fonc.2025.1631490","url":null,"abstract":"<p><strong>Introduction: </strong>Extensive-stage small cell lung cancer (ES-SCLC) is an aggressive malignancy with a poor prognosis. This study aimed to identify and validate clinical and laboratory biomarkers for predicting treatment response and overall survival (OS) in ES-SCLC patients.</p><p><strong>Methods: </strong>We retrospectively analyzed 101 ES-SCLC patients receiving first-line treatment. Logistic and Cox regression analyses identified independent factors influencing treatment efficacy and OS. Subgroup analysis was performed to compare white blood cell (WBC) changes between chemotherapy-alone and chemo-immunotherapy groups. Predictive models were constructed and evaluated via cross-validation, ROC, and calibration curves. Differential expression of key proteins (neuron-specific enolase (NSE), fibrinogen (FIB), and gastrin-releasing peptide precursor (ProGRP)) was validated using GEO database data.</p><p><strong>Results: </strong>Pre-chemotherapy tumor size and post-cycle 2 FIB levels were independent predictors of treatment efficacy. Pre-chemotherapy WBC count, pre-chemotherapy D-dimer, and post-cycle 2 ProGRP were independent risk factors for OS. The predictive models demonstrated strong performance. Subgroup analysis showed no significant difference in WBC changes between treatment regimens (mean change: -2.30 ± 2.47 vs. -2.08 ± 2.45, p=0.659). GEO data confirmed the differential expression of FIB and ProGRP.</p><p><strong>Discussion: </strong>Our findings establish robust and validated models based on readily available clinical metrics (tumor size, WBC, D-dimer, FIB, ProGRP) to predict outcomes in ES-SCLC, which could aid in personalizing treatment strategies. The stability of WBC trends across therapies strengthens the prognostic value of baseline WBC.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1631490"},"PeriodicalIF":3.5,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12460094/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145185238","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Frontiers in OncologyPub Date : 2025-09-11eCollection Date: 2025-01-01DOI: 10.3389/fonc.2025.1550264
Anne H Zur Horst, Steven J M Habraken, Marta Rovituso, Yvonne L B Klaver, Kees H Spruijt, Mischa S Hoogeman
{"title":"Impact assessment of current knowledge gaps and mitigation strategies in clinical FLASH proton therapy through a systematic review.","authors":"Anne H Zur Horst, Steven J M Habraken, Marta Rovituso, Yvonne L B Klaver, Kees H Spruijt, Mischa S Hoogeman","doi":"10.3389/fonc.2025.1550264","DOIUrl":"10.3389/fonc.2025.1550264","url":null,"abstract":"<p><strong>Introduction: </strong>Following first clinical trials, the development of FLASH proton therapy (FLASH-PT) into a mature treatment modality is ongoing, while physical and biological conditions underlying the FLASH effect remain uncertain. Our aim is to assess the impact of these uncertainties on clinical FLASH-PT through a novel approach.</p><p><strong>Methods: </strong>A systematic literature review was conducted to collect relevant <i>in vivo</i> preclinical studies as well as FLASH-PT treatment planning and delivery approaches. This information was used to perform an impact assessment: the FLASH-PT process from patient selection to treatment delivery was divided into steps, and seven FLASH conditions were defined. The FLASH conditions included physical, delivery-related, and radiobiological aspects. For each step and FLASH condition, scores were assigned based on the (i) criticality for clinical applications, (ii) current knowledge, and (iii) available mitigation strategies. These scores were combined to obtain an overall impact for all FLASH conditions ranging from insignificant impact not affecting clinical routine to severe impact causing severe complications for clinical translation.</p><p><strong>Results: </strong>In total, 14 preclinical and 27 treatment planning studies were identified. From these, 47 combined scores were reported in the impact assessment. A severe impact was found for patient selection in the context of radiobiological uncertainties for the robustness of the FLASH effect with respect to beam pauses and interruptions and for the evaluation of dose rate due to their importance in the treatment process combined with remaining unknowns. Moderate to insignificant impact was found for fractionation and FLASH-PT treatment delivery mode (transmission or Bragg peak beams), as these offer strategies to circumvent uncertainties. Overall, dose requirements, the use of multiple fields, and dose rate conditions emerged as the most crucial factors.</p><p><strong>Conclusions: </strong>Since uncertainties about the FLASH conditions hinder the utilization of its full pre-clinical potential in clinical practice, focusing future preclinical experiments to gain further phenomenological rather than only mechanistic insights on these aspects is recommended.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1550264"},"PeriodicalIF":3.5,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12460130/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145185246","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Frontiers in OncologyPub Date : 2025-09-11eCollection Date: 2025-01-01DOI: 10.3389/fonc.2025.1608710
Cong Shen, Haiping Cai, Chengliang Mao, Jiahao Yang, Kai Tang
{"title":"GADD45G as a novel prognostic biomarker and therapeutic target in glioma: integrative analysis of bulk and single-cell RNA sequencing.","authors":"Cong Shen, Haiping Cai, Chengliang Mao, Jiahao Yang, Kai Tang","doi":"10.3389/fonc.2025.1608710","DOIUrl":"10.3389/fonc.2025.1608710","url":null,"abstract":"<p><strong>Background: </strong>Gliomas make up almost half of primary central nervous system tumors. Despite advancements in surgery and neuro-oncology, developing an effective treatment remains challenging. The protein Growth Arrest and DNA-Damage-Inducible, Gamma (GADD45G) is crucial for key cellular functions like DNA repair, genomic stability, and apoptosis. While GADD45G dysregulation has been found in various cancers, its role in glioma is still unclear.</p><p><strong>Methods: </strong>We analyzed unified pan-cancer datasets (TCGA, TARGET, GTEx) from UCSC Xena and integrated glioma data from CGGA and GEO (GSE108476). Prognostic value was assessed via multivariate Cox regression and Kaplan-Meier survival analysis using Gliovis. Single-cell RNA-seq data (GSE103224, GSE138794, GSE173278) were processed with Seurat (R 4.2.2), with Harmony for batch correction and UMAP for visualization. Malignant subclusters were annotated using marker genes. Functional enrichment and cell-type proportion estimation were conducted. Single-cell analysis revealed GADD45G expression patterns and identified its top correlated genes in malignant glioblastoma cells. Overexpression of GADD45G was performed to investigate its impact on cell function. Western blot analysis was used to examine the role of GADD45G in glioma cell invasion and migration.</p><p><strong>Results: </strong>Through comprehensive analysis across multiple datasets, it was found that GADD45G expression is higher in glioma patients compared to normal individuals, and its expression is generally higher in lower-grade gliomas than in glioblastoma. Cox regression analysis indicated that GADD45G has a protective effect. Survival curves further demonstrated that elevated GADD45G levels are associated with improved overall survival in patients. In this study, we identified four highly heterogeneous GBM cell subpopulations using single-cell data. The MES-like cells was significantly associated with poor prognosis. Spearman correlation analysis revealed the correlation between GADD45G and VIM. Further experiments revealed that GADD45G modulates glioma cell invasion and migration, potentially through its effects on EMT-like phenotypic features.</p><p><strong>Conclusion: </strong>GADD45G expression is significantly associated with glioma outcomes and may serve as a promising biomarker for prognosis evaluation. Its involvement in regulating EMT-like phenotypic traits further highlights its therapeutic potential.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1608710"},"PeriodicalIF":3.5,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12460128/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145185263","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Frontiers in OncologyPub Date : 2025-09-11eCollection Date: 2025-01-01DOI: 10.3389/fonc.2025.1597110
Shuheng Shang, Meng Qin, Shuangmei Zhang
{"title":"Long-term complete remission with immunotherapy in advanced RET fusion-positive NSCLC with brain metastases: a case report and literature review.","authors":"Shuheng Shang, Meng Qin, Shuangmei Zhang","doi":"10.3389/fonc.2025.1597110","DOIUrl":"10.3389/fonc.2025.1597110","url":null,"abstract":"<p><p>The rearranged during transfection (RET) fusion is a rare genetic alteration in non-small-cell lung cancer (NSCLC), and the presence of brain metastases significantly influences prognosis. We present a 59-year-old patient diagnosed with RET fusion-positive lung adenocarcinoma which had metastasized to the brain at the time of initial diagnosis, classified as stage IVB, cT3N3M1. Tumor biopsy immunohistochemistry showed PD-L1 positivity (10%). After three cycles of pemetrexed plus cisplatin (PC) regimen combined with camrelizumab (a PD-1 inhibitor), a partial response (PR) was observed through chest computed tomography (CT) and brain magnetic resonance imaging (MRI). The patient underwent whole brain radiotherapy (WBRT) with a total dose of 37.5 Gy over 15 fractions, followed by 3 cycles of the PC regimen plus camrelizumab. Complete remission (CR) was achieved during 30 months of maintenance therapy with pemetrexed plus camrelizumab. The most recent follow-up was in February 2025. Both chest CT and brain MRI continued to show CR, with no clear indications of metastases. During the course of immunochemotherapy, grade 1 bone marrow suppression, but no toxicity of grade 3 or above, was observed. NSCLC patients who have PD-L1 overexpression and RET fusion-positivity may respond well to immunotherapy. Combining radiotherapy with immunotherapy may enhance local control of brain metastases.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1597110"},"PeriodicalIF":3.5,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12460116/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145185391","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Frontiers in OncologyPub Date : 2025-09-11eCollection Date: 2025-01-01DOI: 10.3389/fonc.2025.1647606
Yanjie Yang, Yanyan Liu, Man Yang, Yunli Fan, Wei Du
{"title":"De-escalating adjuvant therapy after pathologic complete response in oral squamous cell carcinoma: Chemoradiotherapy benefits only high-risk subgroups.","authors":"Yanjie Yang, Yanyan Liu, Man Yang, Yunli Fan, Wei Du","doi":"10.3389/fonc.2025.1647606","DOIUrl":"10.3389/fonc.2025.1647606","url":null,"abstract":"<p><strong>Background: </strong>The optimal adjuvant therapy for oral squamous cell carcinoma (SCC) patients achieving pathological complete response (pCR) after neoadjuvant immunochemotherapy (NAIC) remains uncertain. While radiotherapy (RT) and chemoradiotherapy (CRT) improve locoregional control, their comparative efficacy and toxicity profiles in this setting are poorly defined.</p><p><strong>Methods: </strong>Oral SCC patients with pCR post-NAIC were retrospectively enrolled and stratified into RT and CRT groups. Propensity score matching balanced baseline characteristics. Outcomes included 3-year locoregional control (LRC), overall survival (OS), and toxicity. Subgroup analyses evaluated treatment effects by radiologic extranodal extension (rENE) and tumor differentiation.</p><p><strong>Results: </strong>Among 116 patients analyzed (84 matched), CRT showed no significant LRC or OS benefit over RT alone in the overall cohort (LRC: HR 1.89, 95% CI 0.26-4.72, p=0.625; OS: HR 1.45, 95% CI 0.62-3.41, p=0.392). However, subgroup analyses revealed CRT improved outcomes in high-risk patients (rENE+ or poorly differentiated tumors), reducing recurrence by 50% (rENE+: HR 3.12, 95% CI 1.13-8.60, p=0.028; poor differentiation: HR 3.45, 95% CI 1.23-9.68, p=0.019) and enhancing 3-year OS (rENE+: 62.4% <i>vs</i>. 50.1%, p=0.036; poorly differentiated: 68.3% <i>vs</i> 53.8%, HR 2.88, p=0.022). CRT was associated with significantly higher acute and chronic toxicities (Grade 3-5 mucositis: 36.0% <i>vs</i>. 12.1%).</p><p><strong>Conclusion: </strong>CRT should be reserved for high-risk pCR patients (rENE+ or poorly differentiated tumors), while RT alone suffices for low-risk cases. This risk-adapted approach optimizes outcomes while minimizing toxicity.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1647606"},"PeriodicalIF":3.5,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12460139/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145185277","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Frontiers in OncologyPub Date : 2025-09-11eCollection Date: 2025-01-01DOI: 10.3389/fonc.2025.1626529
Cheng Shen, Gen Chen, Zhan Chen, Junjie You, Bing Zheng
{"title":"The risk prediction model for acute urine retention after perineal prostate biopsy based on the LASSO approach and Boruta feature selection.","authors":"Cheng Shen, Gen Chen, Zhan Chen, Junjie You, Bing Zheng","doi":"10.3389/fonc.2025.1626529","DOIUrl":"10.3389/fonc.2025.1626529","url":null,"abstract":"<p><strong>Objective: </strong>One known side effect of transperineal (TP) prostate biopsies is acute urine retention (AUR). We aimed to create and evaluate a predictive model for the post-paracentesis risk of acquiring AUR.</p><p><strong>Methods: </strong>This study included 599 patients undergoing prostate biopsies (April 2020-July 2023) at the Second Affiliated Hospital of Nantong University, selected based on abnormal digital rectal examination and/or PSA (prostate-specificantigen) > 4 ng/mL. Acute urinary retention (AUR) was defined as the inability to void within 72 hours post-biopsy, requiring catheterization. Patients were randomly divided into training (419 cases) and test (180 cases) sets. Univariate logistic analysis and feature selection Boruta and LASSO (Least absolute shrinkage and selection operator) identified predictors, followed by multivariate logistic regression to develop a predictive nomogram for AUR. Internal validation used the test set, with model performance assessed via the c-index, ROC (Receiver Operating Characteristic) curve, calibration plot, and decision curve analysis. The nomogram demonstrated strong discrimination, calibration, and clinical utility for AUR risk prediction.</p><p><strong>Results: </strong>In 86 patients (14.3%), AUR happened. An examination of multivariate logistic regression revealed six distinct risk variables for AUR. Based on these independent risk factors, a nomogram was constructed. The training and validation groups' c-indices showed the model's high accuracy and stability. The calibration curve demonstrates that the corrective effect of the training and verification groups is perfect, and the area under the receiver operating characteristic curve indicates great identification capacity. DCA (Decision Curve Analysis) curves, or decision curve analysis, demonstrated the model's significant net therapeutic effect.</p><p><strong>Discussion: </strong>The nomogram model created in this work can offer a personalized and intuitive analysis of the risk of AUR and has intense discrimination and accuracy. It can help create efficient preventative measures and identify high-risk populations.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1626529"},"PeriodicalIF":3.5,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12460100/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145185377","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Frontiers in OncologyPub Date : 2025-09-11eCollection Date: 2025-01-01DOI: 10.3389/fonc.2025.1673752
Xin Wang, Zhaoyang Qi, Qin Zeng, Dongling Gu, Tianliang Li
{"title":"Radiotherapy for glioma in the AI era: current applications and future prospects.","authors":"Xin Wang, Zhaoyang Qi, Qin Zeng, Dongling Gu, Tianliang Li","doi":"10.3389/fonc.2025.1673752","DOIUrl":"10.3389/fonc.2025.1673752","url":null,"abstract":"<p><p>Gliomas are primary central nervous system tumors characterized by a high recurrence rate and poor prognosis, especially in high-grade forms such as glioblastoma (GBM). Radiotherapy remains a cornerstone in glioma management, particularly following surgical resection. Recent advancements in technology-including intensity-modulated radiotherapy (IMRT), proton therapy, carbon-ion radiotherapy, intraoperative radiotherapy, and ultra-high dose rate FLASH radiotherapy-have improved treatment precision and tumor control. However, clinical challenges persist due to tumor heterogeneity, imaging limitations, and planning variability. In the era of artificial intelligence (AI), novel tools such as radiomics, deep learning, and predictive modeling are increasingly being integrated into glioma radiotherapy workflows. These AI-driven approaches have shown potential to enhance imaging interpretation, automate contouring, optimize treatment planning, and predict clinical outcomes. This review highlights the evolution of glioma radiotherapy, explores the emerging role of AI across various stages of radiotherapy, and discusses future directions for implementing personalized, adaptive, and data-driven strategies in clinical practice.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1673752"},"PeriodicalIF":3.5,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12460113/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145185386","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}