{"title":"Ultrasound-guided percutaneous radiofrequency ablation combined with anti-PD-1 for the treatment of prostate cancer: an experimental study.","authors":"Si Chen, Ruiqing Liu, Shaobo Duan, Beibei Zhang, Yuzhou Wang, Xiaoxiao Li, Yingying Zhao, Zesheng Li, Qi Zhou, Rui Zhang, Linlin Zhang, Xiaoxia Xu, Ru Jang, Juan Zhang, Yaqiong Li, Xiguo Cai, Lianzhong Zhang","doi":"10.3389/fonc.2025.1527763","DOIUrl":"10.3389/fonc.2025.1527763","url":null,"abstract":"<p><strong>Background: </strong>This study seeks to investigate the potential synergistic effects of combining ultrasound-guided percutaneous radiofrequency ablation with anti-PD-1 therapy on prostate cancer, utilizing animal models.</p><p><strong>Methods: </strong>A mouse model of prostate cancer was established by subcutaneous injection of 1 × 10<sup>6</sup> Myc-Cap cells on the right side of FVB mice. When the volume of the tumors reached about 400mm<sup>3</sup>, the mice were randomly divided into four groups and received corresponding intervention treatments. Among them, Group 1 was the blank control group, Group 2 was the simple anti-PD-1 treatment group, Group 3 was the simple radiofrequency ablation group, and Group 4 is the group that received percutaneous radiofrequency ablation combined with anti-PD-1 therapy under ultrasound guidance. The growth of the tumors was observed in mice after treatment in each group, tumor tissues were collected, and the immune status of the mice was analyzed through flow cytometry, immunohistochemistry, immunofluorescence, and other methods.</p><p><strong>Results: </strong>Compared with other treatment groups, ultrasound-guided percutaneous radiofrequency ablation combined with anti-PD-1 therapy significantly reduced the weight and volume of the tumors, demonstrating more effective tumor suppression. At the same time, combination therapy can promote the aggregation of T-cells within the tumor and increase the proportion of cytotoxic T-cells, increase the proportion of M1 macrophages and iNOS expression, and decrease the proportion of M2 macrophages and Arg expression in the local area of the tumors.</p><p><strong>Conclusion: </strong>Local ablation can improve the therapeutic effect of PD-1 monoclonal antibody. Our preliminary results suggest that ultrasound-guided percutaneous radiofrequency ablation, in combination with anti-PD-1 treatment, produces synergistic effects. These effects may be driven by changes in immune cell populations within the tumor's immunosuppressive microenvironment.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1527763"},"PeriodicalIF":3.5,"publicationDate":"2025-03-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11973081/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143802918","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 : 2025-03-24eCollection Date: 2025-01-01DOI: 10.3389/fonc.2025.1511564
Qiangqiang Ge, Zhong Lin, Xuequan Wang, Zhengli Jiang, Yan Hu
{"title":"A seven-LncRNA signature for prognosis prediction of patients with lung squamous cell carcinoma through tumor immune escape.","authors":"Qiangqiang Ge, Zhong Lin, Xuequan Wang, Zhengli Jiang, Yan Hu","doi":"10.3389/fonc.2025.1511564","DOIUrl":"10.3389/fonc.2025.1511564","url":null,"abstract":"<p><strong>Background: </strong>Lung squamous cell carcinoma (LUSC) is a malignant disease associated with poor therapeutic responses and prognosis. Preliminary studies have shown that the dysregulation of long non-coding RNAs (LncRNAs) is linked to cancer development and prognosis. However, research on the role of LncRNAs in LUSC remains limited.</p><p><strong>Methods: </strong>In this study, we aimed to develop a LncRNA signature for improved prognostic prediction in LUSC and to elucidate the underlying mechanisms. We utilized expression data of LncRNAs and clinical information from 471 LUSC patients in The Cancer Genome Atlas (TCGA), randomly dividing them into a training set (n=236) and a testing set (n=235).</p><p><strong>Results: </strong>A prognostic signature model comprising seven LncRNAs was constructed using multivariate Cox regression analysis based on the training set. Using a risk score cutoff value of -0.12 (log2-transformed), patients were categorized into high-risk (n=101) and low-risk (n=370) groups. The high-risk group demonstrated significantly worse overall survival (OS) compared to the low-risk group (p<0.0001). The risk score showed strong prognostic predictive ability for LUSC patients, as evidenced by the area under the ROC curve (AUC: 0.66, 0.67, and 0.67) and nomogram analysis (C-index, calibration, and decision curve analysis) for 1-, 3-, and 5-year survival predictions. Independent prognostic factors for LUSC were identified, including risk group (HR=0.3, 95% CI: 0.22-0.4), stage (HR=1.78, 95% CI: 1.28-2.48), and age (HR=1.02, 95% CI: 1.00-1.04). KEGG enrichment analysis revealed that mRNAs influenced by the seven targeted LncRNAs, associated with immune evasion, were primarily linked to pathways such as chemical carcinogenesis, Th17 cell differentiation, NF-κB signaling, and proteoglycans in cancer. Expression levels of 14 target genes related to tumor immune tolerance were significantly suppressed, with eight confirmed via real-time PCR and western blot analysis. Additionally, CIBERSORT analysis of immune cell-related gene expression between normal and LUSC tissues indicated activation of the immune system in LUSC patients.</p><p><strong>Conclusion: </strong>In conclusion, our findings highlight the clinical significance of the seven LncRNA signature in predicting survival outcomes for LUSC patients.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1511564"},"PeriodicalIF":3.5,"publicationDate":"2025-03-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11973350/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143802955","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 : 2025-03-24eCollection Date: 2025-01-01DOI: 10.3389/fonc.2025.1539362
Nan Jiang, Li-Ping Xu, Fei Li, Pei-Pei Wang, Yuandong Cao
{"title":"Efficacy and safety of simultaneous integrated boost intensity-modulated radiotherapy combined with temozolomide for the postoperative chemotherapy treatment of multifocal high-grade glioma.","authors":"Nan Jiang, Li-Ping Xu, Fei Li, Pei-Pei Wang, Yuandong Cao","doi":"10.3389/fonc.2025.1539362","DOIUrl":"10.3389/fonc.2025.1539362","url":null,"abstract":"<p><strong>Background: </strong>The multifocal manifestation of high-grade glioma is a rare disease with an unfavorable prognosis. The pathogenesis of multifocal gliomas and pathophysiological differences in unifocal gliomas are not fully understood. The optimal treatment for patients with multifocal high-grade glioma is not defined in the current guidelines; therefore, individual case series may be helpful as guidance for clinical decision-making.</p><p><strong>Methods: </strong>Patients with multifocal high-grade glioma treated with simultaneous integrated boost intensity-modulated radiotherapy combined with temozolomide for postoperative treatment at our institution between January 2020 and December 2023 were retrospectively analyzed. Multifocality was neuroradiologically assessed and defined as at least two independent contrast-enhancing foci in the MRI T1 contrast-enhanced sequence. Overall and progression-free survival were calculated from the diagnosis until death and from the start of radiation therapy until the diagnosis of disease progression on MRI for all patients.</p><p><strong>Results: </strong>A total of 42 patients with multifocal high-grade glioma were examined, of which 16 were female and 26 were male. The median age of all patients was 57 years (range: 23-77 years). The median KPS score was 80 (range: 50-100). Complete resection was performed in 10 cases, and partial resection was performed in 32 cases before the start of radiation therapy. The prescription schedule was 54 Gy (1.8 Gy × 30) with an SIB of 60 Gy (2 Gy × 30). Concomitant temozolomide chemotherapy was administered to 40 patients. Median survival was 19 months (95% CI 14.1-23.8 months) and median progression free survival after initiation of RT 13 months (95% CI 9.2-16.7 months). Five patients experienced grade 3 toxicity, none experienced grade 4 toxicity, and no treatment-related deaths occurred.</p><p><strong>Conclusion: </strong>Multifocal high-grade gliomas can be treated safely and efficiently with simultaneous integrated boost intensity-modulated radiotherapy with concomitant and adjuvant TMZ chemotherapy.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1539362"},"PeriodicalIF":3.5,"publicationDate":"2025-03-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11973260/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143802847","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 : 2025-03-24eCollection Date: 2025-01-01DOI: 10.3389/fonc.2025.1526718
Tingjing You, Shengmin Zhang
{"title":"Recent advances in PLGA polymer nanocarriers for ovarian cancer therapy.","authors":"Tingjing You, Shengmin Zhang","doi":"10.3389/fonc.2025.1526718","DOIUrl":"10.3389/fonc.2025.1526718","url":null,"abstract":"<p><p>Ovarian cancer (OC) is the most lethal gynecologic malignancy worldwide, and early diagnosis and effective treatment have been the focus of research in this field. It is because of its late diagnosis, acquired resistance mechanisms, and systemic toxicity of chemotherapeutic agents that the treatment of ovarian cancer is challenging. Combination chemotherapy can potentially improve therapeutic efficacy by activating multiple downstream pathways to overcome resistance and reduce the required dose. In recent years, PLGA-lipid hybrid nanoparticles have demonstrated their potential as an emerging drug delivery system for treating ovarian cancer. PLGA (poly (lactic-co-glycolic acid) has become a highly sought-after biomaterial for the clinical translation of adjustable drug delivery regimens due to its biodegradability, biocompatibility, and multifunctionality, coupled with controlled drug release, which can effectively overcome multidrug resistance and improve the efficiency of chemotherapy. Combination therapies are gradually becoming an ideal alternative to traditional drug formulations. The application of nanoparticles not only improves the therapeutic effect but also reduces the side effects, which provides strong support for personalized precision medicine. We review polymeric nanoparticle carriers for drug combinations used in the treatment of ovarian cancer, particularly the combination of paclitaxel analogs (commonly used first-line therapy for ovarian cancer) with other small molecule therapeutic agents and cavitation combination therapy under ultrasound targeting (<b>Figure 1</b>). The elucidation of these issues will provide a theoretical basis for future exploration of novel NNDDS targeting GRPR for anti-OC therapy. This review presents research on recent advances in PLGA polymer nanoparticles in ovarian cancer, focusing on the use of PLGA degradable microspheres for loading chemotherapeutic agents and ultrasound combination therapy.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1526718"},"PeriodicalIF":3.5,"publicationDate":"2025-03-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11973302/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143802909","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 : 2025-03-24eCollection Date: 2025-01-01DOI: 10.3389/fonc.2025.1513360
Yuxin Ji, Ruonan Li, Guohui Tang, Wenrui Wang, Changjie Chen, Qingling Yang
{"title":"The interrelated roles of RAB family proteins in the advancement of neoplastic growth.","authors":"Yuxin Ji, Ruonan Li, Guohui Tang, Wenrui Wang, Changjie Chen, Qingling Yang","doi":"10.3389/fonc.2025.1513360","DOIUrl":"10.3389/fonc.2025.1513360","url":null,"abstract":"<p><p>Rab Proteins, A Subfamily Of The Ras Superfamily Of Small Gtpases, Are Critical Regulators Of Intracellular Vesicular Trafficking, Which Is Intricately Linked To Various Cellular Processes. These Proteins Play Essential Roles Not Only In Maintaining Cellular Homeostasis But Also In Mediating The Complex Interplay Between Cancer Cells and Their Microenvironment. Rab Proteins Can Act As Either Oncogenic Factors Or Tumor Suppressors, With Their Functions Highly Dependent On The Cellular Context. Mechanistic Studies Have Revealed That Rab Proteins Are Involved In A Variety Of Processes, Including Vesicular Transport, Tumor Microenvironment Regulation, Autophagy, Drug Resistance, and Metabolic Regulation, and Play Either A Promotional Or Inhibitory Role In Cancer Development. Consequently, Targeting Rab Gtpases To Restore Dysregulated Vesicular Transport Systems May Offer A Promising Therapeutic Strategy To Inhibit Cancer Progression. However, It Is Equally Important To Consider The Potential Risks Of Disrupting Rab Functions, As Their Roles Are Highly Context-Dependent and May Have Opposing Effects In Different Malignancies. This Review Focuses On The Multifaceted Involvement Of Rab Family Proteins In Cancer Progression Underscores Their Importance As Potential Therapeutic Targets and Underscores The Need For A Deeper Understanding Of Their Complex Roles In Tumorigenesis.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1513360"},"PeriodicalIF":3.5,"publicationDate":"2025-03-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11974252/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143802914","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 : 2025-03-24eCollection Date: 2025-01-01DOI: 10.3389/fonc.2025.1568568
Lekshmi R Nath, Gautam Sethi, Vijayasteltar B Liju
{"title":"Editorial: Immunomodulatory molecules of natural origin: innovative strategies for combatting cancer.","authors":"Lekshmi R Nath, Gautam Sethi, Vijayasteltar B Liju","doi":"10.3389/fonc.2025.1568568","DOIUrl":"10.3389/fonc.2025.1568568","url":null,"abstract":"","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1568568"},"PeriodicalIF":3.5,"publicationDate":"2025-03-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11973598/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143802770","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}
{"title":"Predictive model using systemic inflammation markers to assess neoadjuvant chemotherapy efficacy in breast cancer.","authors":"Yulu Sun, Yinan Guan, Hao Yu, Yin Zhang, Jinqiu Tao, Weijie Zhang, Yongzhong Yao","doi":"10.3389/fonc.2025.1552802","DOIUrl":"10.3389/fonc.2025.1552802","url":null,"abstract":"<p><strong>Background: </strong>Pathological complete response (pCR) is an important indicator for evaluating the efficacy of neoadjuvant chemotherapy (NAC) in breast cancer. The role of systemic inflammation markers in predicting pCR and the long-term prognosis of breast cancer patients undergoing NAC remains controversial. The purpose of this study was to explore the potential predictive and prognostic value of systemic inflammation markers (NLR, PLR, LMR, NMR) and clinicopathological characteristics in breast cancer patients receiving NAC and construct a pCR prediction model based on these indicators.</p><p><strong>Methods: </strong>A retrospective analysis was conducted on 209 breast cancer patients who received NAC at Nanjing Drum Tower Hospital between January 2010 and March 2020. Independent sample t-tests, chi-square tests, and logistic regression models were used to evaluate the correlation between clinicopathological data, systemic inflammation markers, and pCR. Receiver operating characteristic (ROC) curves were utilized to determine the optimal cut-off values for NLR, PLR, and LMR. Survival analysis was performed using the Kaplan-Meier method and log-rank test. A predictive model for pCR was constructed using machine learning algorithms.</p><p><strong>Results: </strong>Among the 209 breast cancer patients, 29 achieved pCR. During a median follow-up of 68 months, 74 patients experienced local or distant metastasis, and 56 patients died. Univariate logistic regression analysis showed that lymph node status, HER-2 status, NLR, PLR, and LMR were associated with pCR. ROC curve analysis revealed that the optimal cut-off values for NLR, PLR, and LMR were 1.525, 113.620, and 6.225, respectively. Multivariate logistic regression analysis indicated that lymph node status, NLR, and LMR were independent predictive factors for pCR. Survival analysis demonstrated that lymph node status, NLR, and LMR were associated with prognosis. Machine learning algorithm analysis identified the random forest (RF) model as the optimal predictive model for pCR.</p><p><strong>Conclusion: </strong>This study demonstrated that lymph node status, NLR, and LMR had significant value in predicting pCR and prognosis in breast cancer patients. The RF model provides a simple and cost-effective tool for pCR prediction, offering strong support for clinical decision-making in breast cancer treatment and aiding in the optimization of individualized treatment strategies.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1552802"},"PeriodicalIF":3.5,"publicationDate":"2025-03-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11973675/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143802898","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 : 2025-03-21eCollection Date: 2025-01-01DOI: 10.3389/fonc.2025.1509619
Shuai Sun, Xinyue Gong, Yongguang Liang, Yuliang Sun, Dan Que, Yangchun Xie, Shumeng He, Lei He, Hao Liang, Yijun Wang, Xinyi Wu, Cheng Wang, Bo Yang, Jie Qiu, Ke Hu, Fuquan Zhang
{"title":"Evaluating the Implementation of fan-beam CT-guided online adaptive re-planning in definitive cervical cancer radiotherapy.","authors":"Shuai Sun, Xinyue Gong, Yongguang Liang, Yuliang Sun, Dan Que, Yangchun Xie, Shumeng He, Lei He, Hao Liang, Yijun Wang, Xinyi Wu, Cheng Wang, Bo Yang, Jie Qiu, Ke Hu, Fuquan Zhang","doi":"10.3389/fonc.2025.1509619","DOIUrl":"10.3389/fonc.2025.1509619","url":null,"abstract":"<p><strong>Background: </strong>This study aims to investigate the feasibility of fan-beam computed tomography (FBCT)-guided online adaptive radiotherapy (oART) in radical radiotherapy for cervical cancer.</p><p><strong>Methods: </strong>Ten patients who underwent radical radiotherapy for cervical cancer were enrolled in this study. All patients received external beam radiation therapy (EBRT) with a prescription dose of 50.4 Gy/28f, and daily oART with FBCT guidance was performed. Dosimetric analysis was conducted on 278 fractions, comparing the adaptive and scheduled plans. The γ passing rate was measured through <i>in-vivo</i> dose monitoring during treatment, using a 3%/3mm gamma criterion with an 88% threshold for alerts. The time invested in the oART workflow was recorded at each step. Acute toxicities were classified following the Common Terminology Criteria for Adverse Events (CTCAE) version 5.0.</p><p><strong>Results: </strong>The adaptive plans demonstrated a dosimetric advantage in target coverage and/or organs at risk (OARs) sparing across all 278 fractions. Compared to the scheduled plan, the adaptive plan showed improved dose received by 95% (D95) of planning target volume (PTV), conformity index (CI), and homogeneity index (HI) (P<0.001). Among the three PTVs, the PTV of uterus (PTV_U) benefited most from dosimetric improvements in the adaptive plan, followed by the PTV of cervix, vagina, and parametrial tissues (PTV_C), while the PTV of lymph node (PTV_N) exhibited the least enhancement. For OARs, the adaptive plan achieved reductions in the dose to the most irradiated 2 cm³ volume (D2cc) for the rectum, bladder, and small intestine (P<0.001). For patients with ovarian conservation, the dose to the 50% volume (D50) and the mean dose of the bilateral ovaries were decreased (P<0.001). The mean γ passing rate across all fractions was 99.24%. The mean duration of the oART workflow was 22.82 ± 3.61 min, with auto-segmentation & review (44.40%) and plan generation & evaluation (22.02%) being the most time-intensive steps. The incidence of Grade 1-2 acute non-hematological toxicity was 60%, with no cases of Grade 3 or higher observed.</p><p><strong>Conclusions: </strong>The implementation of FBCT-guided oART in radical radiotherapy for cervical cancer was feasible. This approach has shown significant improvements in dose distribution and the potential to provide clinical benefits by reducing acute toxicity.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1509619"},"PeriodicalIF":3.5,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11968674/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143794853","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 : 2025-03-21eCollection Date: 2025-01-01DOI: 10.3389/fonc.2025.1542955
Hengyu Tian, Qinghua He, Chidan Wan
{"title":"Conversion therapy combined with ALPPS for the treatment of intrahepatic cholangiocarcinoma: a case report.","authors":"Hengyu Tian, Qinghua He, Chidan Wan","doi":"10.3389/fonc.2025.1542955","DOIUrl":"10.3389/fonc.2025.1542955","url":null,"abstract":"<p><strong>Rationale: </strong>Intrahepatic cholangiocarcinoma (ICC) is a highly malignant liver tumor with limited treatment options for advanced cases. Conversion therapy combining immunotherapy, targeted therapy, and chemotherapy offers a promising approach to enable surgical resection, which remains the only curative option.</p><p><strong>Patient concerns: </strong>A 67-year-old male presented with right upper abdominal pain for two months. Imaging and biopsy confirmed advanced ICC (Stage IV), with a 98 mm tumor, lymphadenopathy, and elevated tumor markers (CA199: 1190.4 U/ml). The disease was deemed unresectable.</p><p><strong>Diagnosis: </strong>The patient was diagnosed with advanced ICC involving a large hepatic mass, lymph node metastasis, and insufficient liver reserve for conventional resection.</p><p><strong>Interventions: </strong>The patient received six months of oxaliplatin plus gemcitabine (GEMOX), lenvatinib, and toripalimab, achieving significant tumor regression. A two-step ALPPS procedure was then performed, comprising portal vein ligation and right hepatectomy.</p><p><strong>Outcomes: </strong>The treatment reduced tumor size (98 mm to 60 mm), normalized tumor markers, and improved liver reserve. Postoperative pathology confirmed >80% tumor remission with negative margins. At 12 months post-surgery, the patient remained disease-free.</p><p><strong>Lessons: </strong>This case demonstrates that advanced ICC can be downstaged with systemic therapy, enabling resection via ALPPS. The combination of GEMOX, lenvatinib, and toripalimab is an effective and safe conversion therapy regimen. This approach may serve as a model for managing similar advanced cases.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1542955"},"PeriodicalIF":3.5,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11968688/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143794892","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 : 2025-03-21eCollection Date: 2025-01-01DOI: 10.3389/fonc.2025.1551937
Julien Haemmerli, Samuel Khatchatourov, Etienne Chaboudez, Leonard Roth, Abiram Sandralegar, Insa Janssen, Denis Migliorini, Karl Schaller, Philippe Bijlenga
{"title":"Surgical and clinical impacts of mixed reality-guided glioblastoma resection versus standard neuronavigation: improving tumor surgery.","authors":"Julien Haemmerli, Samuel Khatchatourov, Etienne Chaboudez, Leonard Roth, Abiram Sandralegar, Insa Janssen, Denis Migliorini, Karl Schaller, Philippe Bijlenga","doi":"10.3389/fonc.2025.1551937","DOIUrl":"10.3389/fonc.2025.1551937","url":null,"abstract":"<p><strong>Background: </strong>Glioblastomas (GBM) are typically treated with surgery and radio-chemotherapy, with patient survival often depending on the extent of tumor resection. This study compares outcomes of GBM surgery using 5-ALA, intraoperative neuroelectrophysiology, and neuro-navigation, either in a standard setting (STD) or enhanced by mixed reality (MR) guidance.</p><p><strong>Methods: </strong>This retrospective study included GBM patients who underwent resection at Geneva University Hospitals between 2015 and mid-2022, excluding biopsies and partial debulking. Primary outcomes included postoperative residual tumor volume (RV) based on postoperative contrast uptake on the MRI, while secondary outcomes were gross total resection (GTR), extent of resection (EOR), new postoperative deficits, overall survival (OS), progression-free survival (PFS), and Karnofsky performance scores. Confounding factors such as intraoperative monitoring and use of fluorescence were analyzed.</p><p><strong>Results: </strong>Of 115 patients, 76 were in the STD group and 39 in the MR group, with comparable demographics. The MR group had significantly lower RV (median 0.01 cm³ vs. 0.34 cm³, p=0.008) and higher GTR rates (median 50% vs. 26.7%). EOR was also superior in the MR group (median 99.9% vs. 98.2%, p=0.002). New focal deficits occurred in 39% (STD) and 36% (MR) of cases (p=0.84). While median OS was not significantly different (475 vs. 375 days, p=0.63), median PFS was longer in the MR group (147 vs. 100 days, p=0.004).</p><p><strong>Conclusion: </strong>MR guidance improves the quality of tumor resection and enhances progression-free survival without increasing postoperative deficits, although it does not significantly impact overall survival.</p>","PeriodicalId":12482,"journal":{"name":"Frontiers in Oncology","volume":"15 ","pages":"1551937"},"PeriodicalIF":3.5,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11968386/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143795134","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}