{"title":"Predictive significance of MPT-driven necrosis-related genes signature in gastric cancer and their impact on the tumor microenvironment.","authors":"Silan Huang, Lingli Huang, Qi Jiang, Chang Jiang, Guifang Guo","doi":"10.1007/s12094-024-03832-7","DOIUrl":"10.1007/s12094-024-03832-7","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer (GC) presents significant management challenges. MPT-driven necrosis (MPTDN) plays a significant role in various conditions, but its connection with GC is unclear. This study aimed to investigate the predictive significance of MPTDN-related genes (MPTDNRGs) in GC and their effect on the tumor immune microenvironment (TIME).</p><p><strong>Methods: </strong>RNA sequencing data for GC were sourced from TCGA and GEO databases. The mutation profiles and MPTDNRG expression between tumor and normal samples were assessed. Prognostic mRNAs were identified using univariate Cox regression and LASSO regression. GC patients were classified into high- and low-risk groups according to risk scores, followed by survival analysis and evaluation of correlations between MPTDN score and clinicopathological features, functional pathway, TIME, and responses to immunotherapy.</p><p><strong>Results: </strong>MPTDNRGs exhibited a 64% mutation rate in GC, with 22 showing significant expression differences. Univariate Cox and LASSO regression identified 15 independently prognostic MPTDNRGs. The prognostic risk model stratified patients into two groups, revealing significant differences in overall and disease-free survival. A nomogram incorporating the signature and clinical characteristics showed strong specificity and sensitivity in predicting prognosis. The MPTDN score was significantly associated with clinical characteristics, functional pathways, and TIME. scRNA-seq analysis indicated higher MPTDN-signature expression in CD8 + T cells, malignant cells, and myofibroblasts. TIDE analysis suggested high-risk patients have reduced responses to immunotherapy, while low-risk patients could benefit more. Importantly, validation using urothelial carcinoma data confirmed a better prognosis for low-risk patients with immunotherapy.</p><p><strong>Conclusion: </strong>This study highlights the importance of MPTDN-related signatures in predicting GC prognosis and guiding therapeutic decisions.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":" ","pages":"3015-3028"},"PeriodicalIF":2.8,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142848332","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Targeting mitochondria and programmed cell death as potential interventions for metastatic castration-resistant prostate cancer.","authors":"Amonlaya Amantakul, Akara Amantakul, Suwalee Pojchamarnwiputh, Nipon Chattipakorn, Siriporn Chaisin Chattipakorn, Jirapas Sripetchwandee","doi":"10.1007/s12094-024-03784-y","DOIUrl":"10.1007/s12094-024-03784-y","url":null,"abstract":"<p><p>Prostate cancer is one of the major causes of morbidity and mortality in men worldwide. Most patients with prostate cancer will turn into end-of-life stage when those tumor cells become metastatic castration-resistant prostate cancer (mCRPC). The mCRPC subsequently developed a resistance to androgen signaling. The current regimens for mCRPC therapy are still ineffective. Much evidence from in vitro and in vivo studies explored the roles of therapeutic interventions targeted at the mitochondria and programmed cell death for prostate cancer therapy. The present review will focus on the recent medications which targeted at mitochondria and programmed cell death in mCRPC and the significant findings from each study will be summarized and discussed. Development of therapeutic interventions, particularly at mitochondrial and cytotoxic targets for treatment of mCRPC without inducing cellular toxicity of normal tissues will be considered as the novel therapeutic strategy for mCRPC.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":" ","pages":"2852-2875"},"PeriodicalIF":2.8,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142840061","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Nicolas Feltes Benitez, Joan Lozano, Carlos G Forero, Montserrat Colomer I Truyols, Saturio Paredes Rubio, Esther Jovell-Fernandez
{"title":"PSA bounce: understanding temporal fluctuations in prostate cancer after external radiotherapy.","authors":"Nicolas Feltes Benitez, Joan Lozano, Carlos G Forero, Montserrat Colomer I Truyols, Saturio Paredes Rubio, Esther Jovell-Fernandez","doi":"10.1007/s12094-024-03816-7","DOIUrl":"10.1007/s12094-024-03816-7","url":null,"abstract":"<p><strong>Purpose: </strong>Prostate-specific antigen (PSA) bounce is a transient elevation in PSA levels commonly observed after radiotherapy. This study aims to investigate the characteristics, timing, and clinical implications of PSA bounce (PSA-B) in prostate cancer patients treated with external beam radiotherapy (EBRT), exploring potential causes and its relevance in patient management.</p><p><strong>Materials and methods: </strong>Between 2013 and 2019, 629 patients with localized prostate cancer were treated with EBRT. After excluding patients with fewer than four PSA measurements or follow-up under 3 years (n = 184), 445 patients were analyzed. The median follow-up duration was 5.9 years (36-105 months). PSA-B was defined as a rise of ≥ 0.2 ng/mL above the nadir, followed by a subsequent decline to or below the nadir. PSA relapse was defined according to Phoenix definition.</p><p><strong>Results: </strong>A total of 64 patients (14.4%) experienced PSA-B at a median of 31 months (6-68 months). Univariable analysis identified age (p < 0.001), risk group (p < 0.001), perineural invasion (p < 0.007), radiotherapy duration (p < 0.001), and the absence of concurrent hormonal therapy (p < 0.001) as independent predictors of PSA-B. Multivariable analysis confirmed age and high-risk group as significant factors. PSA relapse occurred in 10.3% of cases, with only one patient who experienced both PSA-B and relapse.</p><p><strong>Conclusions: </strong>PSA-B is a common phenomenon in localized prostate cancer patients post-EBRT. Factors such as age, risk group, perineural invasion, radiotherapy duration, and hormonal treatment use are associated with PSA-B occurrence. Understanding its mechanisms is crucial for optimizing prostate cancer management.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":" ","pages":"3154-3162"},"PeriodicalIF":2.8,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142866167","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Influence of melatonin supplementation on cancer-related fatigue: a meta-analysis of randomized controlled trials.","authors":"Yongchao Li, Wencai Zhang, Xiaochun Zeng, Lu Zhou, Wenjuan He, Yadong Peng","doi":"10.1007/s12094-024-03824-7","DOIUrl":"10.1007/s12094-024-03824-7","url":null,"abstract":"<p><strong>Purpose: </strong>Previous studies that evaluated the influence of melatonin supplementation on cancer-related fatigue (CRF) revealed inconsistent results. The present meta-analysis was performed to systematically evaluate the influence of melatonin on the severity of fatigue in patients with cancer.</p><p><strong>Methods: </strong>Relevant randomized controlled trials (RCTs) were acquired by conducting a comprehensive search in the PubMed, Embase, and Cochrane Library databases. Only RCTs published as full-length English-language articles were included. A random-effects model was utilized to combine the findings by incorporating its potential influence.</p><p><strong>Results: </strong>Nine RCTs were included for the meta-analysis. Compared to the placebo, melatonin supplementation improved the symptoms of fatigue of these patients (standardized mean difference [SMD]: -0.23, 95% confidence interval [CI]: -0.44 to -0.01, p = 0.04, I<sup>2</sup> = 53%). The univariate analysis suggested that the treatment duration was significantly correlated with the improvement of melatonin supplementation on CRF (coefficient = -0.0063, p = 0.02), which largely explains the source of heterogeneity (adjusted R<sup>2</sup> = 83.7%). The subgroup analysis revealed significantly improved fatigue in studies with treatment durations of ≥13 weeks, but not in studies with treatment durations of <13 weeks (SMD: -0.38 vs. 0.06, p for subgroup difference = 0.02). The further subgroup analysis suggested that the results were not significantly influenced by the type of cancer, status (advanced cancer or overall cancer), sample size, treatment (active anticancer treatment or palliative care only), dose of melatonin, or scale for evaluating fatigue symptoms.</p><p><strong>Conclusions: </strong>Melatonin supplementation may relieve CRF, especially for intervention durations of ≥13 weeks.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":" ","pages":"3232-3244"},"PeriodicalIF":2.8,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903940","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Identification of a gene score related to antigen processing and presentation machinery for predicting prognosis in head and neck squamous cell carcinoma and its potential implications for immunotherapy.","authors":"Xue-Liang Fang, Qing-Jie Li, Li Wang, Yu-Xuan Shi, Li-Ya Hu, Xuan-Yu Zhao, Wei Lv, Hong-Meng Yu","doi":"10.1007/s12094-024-03829-2","DOIUrl":"10.1007/s12094-024-03829-2","url":null,"abstract":"<p><strong>Background: </strong>Despite its crucial role in immune surveillance and cell survival of tumors, the significance of MHC antigen processing and presentation machinery (APM) is still not fully understood in head and neck squamous cell carcinoma (HNSCC). We sought to develop an APM gene score (APMGS) to predict prognosis and reveal the molecular and immune traits of the APMGS-defined subgroups in HNSCC.</p><p><strong>Methods: </strong>Based on the APM-related genes acquired from 6 databases, 117 combined machine learning algorithms were applied to develop APMGS with The Cancer Genome Atlas (TCGA)-HNSCC database and validated with the Gene Expression Omnibus (GEO) dataset. Comprehensive analysis was performed to investigate the molecular and immune features of APMGS subgroups.</p><p><strong>Results: </strong>The APMGS constructed by StepCox [both] + Ridge method achieved the highest C-index and area under curve (AUC) at 3 years and were thus adopted as the final model. Low-APMGS patients exhibited superior overall survival compared with high-APMGS patients in both TCGA and GEO cohorts. Subsequent analysis confirmed that a low APMGS was associated with immune response-related pathways; low TP53 mutation rate and low tumor mutation burden (TMB); a less aggressive phenotype; high infiltration of activated CD4<sup>+</sup> memory T cells, CD8<sup>+</sup> T cells, follicular helper T cells, and Tregs; active immunity; and higher sensitivity to chemotherapeutic and targeted agents. In contrast, a high APMGS linked to proteasome and protein export pathways; high TP53 mutation rate and high TMB; a more aggressive phenotype; high infiltration of M0 macrophages and eosinophils; suppressed immunity; and lower sensitivity to chemotherapeutic and targeted agents.</p><p><strong>Conclusions: </strong>Our findings suggest that APMGS has potential to predict the prognosis, and molecular and immune characteristics of HNSCC, and may also serve as an indicator for immunotherapy benefit.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":" ","pages":"3204-3222"},"PeriodicalIF":2.8,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142911065","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Autologous tumor lysate-loaded dendritic cell vaccination in glioblastoma patients: a systematic review of literature.","authors":"Siddharth Shah, Aiswarya Nag, Brandon Lucke-Wold","doi":"10.1007/s12094-024-03830-9","DOIUrl":"10.1007/s12094-024-03830-9","url":null,"abstract":"<p><p>Glioblastoma (GBM) is one of the most common primary malignant brain tumors. Annually, there are about six instances recorded per 100,000 inhabitants. Treatment for GB has not advanced all that much. Novel medications have been investigated recently for the management of newly diagnosed and recurring instances of GBM. For GBM, surgery, radiation therapy, and alkylating chemotherapy are often used therapies. Immunotherapies, which use the patient's immune reaction against tumors, have long been seen as a potential cancer treatment. One such treatment is the dendritic cell (DC) vaccine. This cell-based vaccination works by stimulating the patient's own dendritic cells' antigenic repertoire, therefore inducing a polyclonal T-cell response. Systematic retrieval of information was performed on PubMed, Embase, and Google Scholar. Specified keywords were used to search, and the articles published in peer-reviewed scientific journals were associated with brain GBM, cancer, and Autologous Tumor Lysate-Loaded Dendritic Cell Vaccination. Selected 90 articles were used in this manuscript, of which 30 articles were clinical trials. Compared to shared tumor antigen peptide vaccines, autologous cancer DCs have a greater ability to stimulate the immune system, which is why dendritic cell fusion vaccines have shown early promise in several clinical studies. Survival rates for vaccinated patients were notably better compared to matched or historical controls. For newly diagnosed patients, the median overall survival (mOS) ranged from 15 to 41.4 months, while the progression-free survival (PFS) ranged from 6 to 25.3 months. We discovered through this analysis that autologous multiomics analysis of DC vaccines showed enhanced antitumor immunity with a focus on using activated, antigen-loaded donor DCs to trigger T-cell responses against cancer, particularly in glioblastoma. It also showed improved patient survival, especially when combined with standard chemoradiotherapy. DC vaccines show promise in treating GBM by enhancing survival and reducing tumor recurrence. However, challenges in vaccine production, antigen selection, and tumor heterogeneity highlight the need for continued research and optimization to improve efficacy and patient outcomes.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":" ","pages":"2889-2903"},"PeriodicalIF":2.8,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142878429","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Camilly Victória Campanharo, Lívia Valle Dos Santos Silveira, Débora Dummer Meira, Matheus Correia Casotti, Lorena Souza Castro Altoé, Iúri Drumond Louro, André Felipe Monteiro Gonçalves, André Manhães Machado, Breno Sousa Paiva, Ester de Souza Inocencio, Fabio Victor Vieira Rocha, Fellipe Pesente, Giulia de Souza Cupertino de Castro, João Pedro Dos Santos da Paixão, José Henrique Borges Bourguignon, Júlia Salarini Carneiro, Juliana Ribeiro de Oliveira, Pâmela de Souza Freire, Sophia Bridi Zamprogno, Taissa Dos Santos Uchiya, Thais de Paula Rezende, Vinícius de Pádua Sanders Medeiros
{"title":"Pan-cancer and multiomics: advanced strategies for diagnosis, prognosis, and therapy in the complex genetic and molecular universe of cancer.","authors":"Camilly Victória Campanharo, Lívia Valle Dos Santos Silveira, Débora Dummer Meira, Matheus Correia Casotti, Lorena Souza Castro Altoé, Iúri Drumond Louro, André Felipe Monteiro Gonçalves, André Manhães Machado, Breno Sousa Paiva, Ester de Souza Inocencio, Fabio Victor Vieira Rocha, Fellipe Pesente, Giulia de Souza Cupertino de Castro, João Pedro Dos Santos da Paixão, José Henrique Borges Bourguignon, Júlia Salarini Carneiro, Juliana Ribeiro de Oliveira, Pâmela de Souza Freire, Sophia Bridi Zamprogno, Taissa Dos Santos Uchiya, Thais de Paula Rezende, Vinícius de Pádua Sanders Medeiros","doi":"10.1007/s12094-024-03819-4","DOIUrl":"10.1007/s12094-024-03819-4","url":null,"abstract":"<p><p>The pan-cancer and multi-omics approach is motivated by the genetic and molecular complexity inherent in the varied types of cancer. This method presents itself as a crucial resource for advancing early diagnosis, defining prognoses and identifying treatments that share common bases between different forms of tumors. The aim of this article is to explore pan-cancer analysis in conjunction with multi-omics strategies, evaluating laboratory, computational, clinical procedures and their consequences, as well as examining the tumor microenvironment, epigenetics and future directions of these technologies in patient management. To this end, a literature review was conducted using PUBMED, resulting in the selection of 260 articles, of which 81 were carefully chosen to support this analysis. The pan-cancer methodology is applied to the study of this microenvironment with the aim of investigating its common characteristics through multiomics data. The development of new therapies depends on understanding the oncogenic pathways associated with different cancers. Thus, the integration of multi-omics and pan-cancer analyzes offers an innovative perspective in the search for new control points, metabolic pathways and markers, in addition to facilitating the identification of patterns common to multiple cancer types, allowing the development of targeted treatments. In this way, the convergence of multiomics and clinical approaches promotes a broad view of cancer biology, leading to more effective and personalized therapies.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":" ","pages":"2936-2954"},"PeriodicalIF":2.8,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142900170","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yang Yang, Xiao Ma, Yue Li, Lihua Jin, Xianchun Zhou
{"title":"The evolving tumor-associated adipose tissue microenvironment in breast cancer: from cancer initiation to metastatic outgrowth.","authors":"Yang Yang, Xiao Ma, Yue Li, Lihua Jin, Xianchun Zhou","doi":"10.1007/s12094-024-03831-8","DOIUrl":"10.1007/s12094-024-03831-8","url":null,"abstract":"<p><p>Adipocytes represent a significant proportion of breast tissue, comprising between 3.7 and 37% of stromal tissue. They play a pivotal role in metabolic regulation, energy supply, metabolic regulation, support effects, and cytokine release within the breast. In breast cancer (BC) tissue, adipocytes engage in intricate crosstalk with BC cells, playing a key role in tumor proliferation, invasion, metastasis formation, and metabolic remodeling. This is due to the provision of hormones, adipokines, and fatty acids to tumor cells by the adipocytes. With the initiation of metastatic outgrowth of BC, the peritumoral adipose tissue exhibits abundant and intricate changes based on its original construction and function, which convert it into a tumor-associated adipose tissue microenvironment (TAAME). It includes some specific adipocytes: adipose-derived stem cells (ASCs), cancer-associated adipocytes (CAAs), adipocyte-derived fibroblasts (ADFs), etc. From a mechanistic standpoint, specific adipocytes can facilitate the proliferation, invasion, metastasis, and angiogenesis of BC cells by secreting a multitude of cytokines (IL-6) and adipokines (leptin), which collectively create an environment conducive to BC progression. It is of paramount importance to recognize the TAAME as a crucial target for the diagnosis, treatment, and drug resistance of BC. Consequently, the review presents an overview of the characteristics and interactions of specific adipocytes within TAAME cell populations. This will facilitate the development of more effective personalized therapies against BC progression, relapse, and metastasis.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":" ","pages":"2778-2788"},"PeriodicalIF":2.8,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142886406","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Association of tumor treating fields (TTFields) therapy with overall survival in newly diagnosed glioblastoma.","authors":"Siddharth Shah, Aiswarya Nag, Brandon Lucke-Wold","doi":"10.1007/s12094-025-03849-6","DOIUrl":"10.1007/s12094-025-03849-6","url":null,"abstract":"<p><p>Glioblastoma (GBM) is a rare and aggressive primary central nervous system tumor with high morbidity and mortality. Standard treatments include surgery, radiotherapy (RT), and temozolomide (TMZ) chemotherapy, but these options are often insufficient and cause severe side effects. Tumor Treating Fields (TTFields) have emerged as a fourth treatment, offering improved survival, better prognosis, and minimal side effects, significantly enhancing the quality of life for GBM patients. For newly diagnosed cases, TTFields combined with TMZ is the recommended standard of care for eligible patients. Current GBM therapy focuses on extending survival while reducing harm. Ongoing research seeks to explore TTFields in innovative radiological-based therapeutic paradigms.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":" ","pages":"2904-2912"},"PeriodicalIF":2.8,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143076467","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Angel Montero, Ovidio Hernando, Mercedes López, Jeannette Valero, Raquel Ciérvide, Emilio Sánchez, Alejandro Prado, Helena B Zobec, Xin Chen-Zhao, Beatriz Álvarez, Mariola García-Aranda, Leyre Alonso, Rosa Alonso, Pedro Fernández-Letón, Carmen Rubio
{"title":"SABR tolerance after prostatectomy: pushing the boundaries of ultrahypofractionation.","authors":"Angel Montero, Ovidio Hernando, Mercedes López, Jeannette Valero, Raquel Ciérvide, Emilio Sánchez, Alejandro Prado, Helena B Zobec, Xin Chen-Zhao, Beatriz Álvarez, Mariola García-Aranda, Leyre Alonso, Rosa Alonso, Pedro Fernández-Letón, Carmen Rubio","doi":"10.1007/s12094-025-03845-w","DOIUrl":"10.1007/s12094-025-03845-w","url":null,"abstract":"<p><strong>Objective: </strong>To evaluate the feasibility and tolerance of ultra-hypofractionated SABR (stereotactic ablative radiation therapy) protocol following radical prostatectomy.</p><p><strong>Patients and methods: </strong>We included patients undergoing adjuvant or salvage SABR between April 2019 and April 2023 targeting the surgical bed and pelvic lymph nodes up to a total dose of 36.25 Gy (7.25 Gy/fraction) and 26 Gy (5.2 Gy/fraction), respectively, in 5 fractions on alternate days with an urethra sparing protocol. Acute and late adverse effects were assessed using the CTCAE v5.0. Pearson's chi-square test for categorical variables was used to compare characteristics and possible associations among different subgroups.</p><p><strong>Results: </strong>Adjuvant radiation therapy (ART) was administered to 40 high-risk patients (detectable post-surgery PSA, Grade Group 4/5, nodal involvement, R1/R2 resection margin), while salvage radiotherapy (SRT) was delivered to 60 patients with rising PSA levels post-undetectable values. Elective nodal irradiation was performed in 57 patients, with 11 additional patients receiving a simultaneous integrated boost (total dose: 40 Gy in 5 fractions) for macroscopic nodal disease. Twenty-four high-risk patients underwent 24-months androgen deprivation therapy (ADT). Treatment was well-tolerated with minimal toxicity. The maximum grade of SABR-related toxicity observed was grade 3. Acute gastrointestinal (GI) toxicity included seven cases of grade 2 and one of grade 3, while acute genitourinary (GU) events were limited to grade 2 in eight patients. Early-late toxicity included two cases of grade 3 and seven of grade 2 for GI, and 11 cases of grade 2 for GU. No toxicity above grade 3 was reported. With a median follow-up of 24 months (6-60 months), 14 patients experienced disease recurrence.</p><p><strong>Conclusions: </strong>Ultra-hypofractionated adjuvant/salvage SABR appears feasible and safe. Longer follow-up is needed to validate observed outcomes.</p>","PeriodicalId":50685,"journal":{"name":"Clinical & Translational Oncology","volume":" ","pages":"3144-3153"},"PeriodicalIF":2.8,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143043287","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}