R Luke Bradley, Edwin Paul, Sharda Singh, Thomas E Hutson
{"title":"CD70-targeted allogeneic CAR T-cell therapy for clear cell renal cell carcinoma.","authors":"R Luke Bradley, Edwin Paul, Sharda Singh, Thomas E Hutson","doi":"10.1080/14737140.2025.2548489","DOIUrl":"10.1080/14737140.2025.2548489","url":null,"abstract":"<p><strong>Introduction: </strong>Allogeneic chimeric antigen receptor (CAR) T-cell therapy is a promising yet underexplored treatment for clear cell renal cell carcinoma (ccRCC), potentially more effective than existing treatment options. This review compares several ongoing preclinical and clinical trials using CAR T-cell therapy.</p><p><strong>Areas covered: </strong>This review discusses the development of CAR T-cell therapy in ccRCC, covering four significant themes: (1) optimizing therapeutic efficacy through combination strategies, (2) the translation pathway from preclinical development to clinical application, (3) safety and toxicity management, and (4) immune response modulation in the tumor microenvironment. Finally, this review highlights opportunities to overcome current limitations and guide future therapeutic approaches. We conducted a structured review of existing research using the PubMed and Google Scholar databases from the past 5 years, compiled relevant studies on CAR T-cell therapies for ccRCC, and categorized them into four key themes, which were then cross-analyzed to identify trends, challenges, and emerging limitations.</p><p><strong>Expert opinion: </strong>Development of universal CAR T-cells may be more affordable, more accessible, and easier to administer in less time with fewer mechanical failures than autologous CAR T-cell therapy. Some challenges persist, including patient toxicities, depletion of CAR T-cells in vivo, and an immunosuppressive tumor microenvironment.</p>","PeriodicalId":12099,"journal":{"name":"Expert Review of Anticancer Therapy","volume":" ","pages":"1-10"},"PeriodicalIF":2.8,"publicationDate":"2025-08-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144845055","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":"The relationship between statin use and breast cancer risk: NHANES 2003-2016 and Mendelian randomization study.","authors":"Hao Shi, Ting Li, Yan Xu","doi":"10.1080/14737140.2025.2548488","DOIUrl":"10.1080/14737140.2025.2548488","url":null,"abstract":"<p><strong>Background: </strong>Breast cancer is a prevalent malignancy, and statins are commonly used in the treatment of hyperlipidemia. However, the association between the use of statins and the risk of breast cancer is unclear. The aim of our study was to explore the relationship between statins and breast cancer risk by combining data analysis from the NHANES (National Health and Nutrition Examination Survey) and a Mendelian randomization (MR) study.</p><p><strong>Research design and methods: </strong>We collected and compiled data from the NHANES between 2003 and 2016. Logistic regression models were used to evaluate the associations between statin use and the risk of breast cancer. To further validate our findings, we selected two sets of instrumental variables and conducted a MR study. Additionally, we evaluated the robustness and reliability of the MR results through sensitivity analyses.</p><p><strong>Results: </strong>Based on the results of multivariate logistic regression analysis, statins may be a potential protective factor against breast cancer. For the MR analyses, inverse-variance weighted (IVW) analysis also revealed an association between exposure to statin-targeted inhibition and the risk of breast cancer.</p><p><strong>Conclusion: </strong>The administration of statins contributes to reduce the risk of breast cancer, which may open up new perspectives on breast cancer prevention.</p>","PeriodicalId":12099,"journal":{"name":"Expert Review of Anticancer Therapy","volume":" ","pages":"1-11"},"PeriodicalIF":2.8,"publicationDate":"2025-08-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144854971","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}
Jiaxin Huang, Jiaqi Tan, Nanfeng Meng, Junrong Wang, Peng Han, Hang Wang
{"title":"The transcription factor HOXB7 significantly enhances the expression of PIGT through the Wnt/β-catenin signaling pathway, thereby promoting the proliferation and deterioration of HCC.","authors":"Jiaxin Huang, Jiaqi Tan, Nanfeng Meng, Junrong Wang, Peng Han, Hang Wang","doi":"10.1080/14737140.2025.2544864","DOIUrl":"https://doi.org/10.1080/14737140.2025.2544864","url":null,"abstract":"<p><strong>Background: </strong>Glycosylphosphatidylinositol-anchored proteins (GPI-APs) contribute to cancer progression, with their glycolipid modification mediated by glycosylphosphatidylinositol transamidase (GPIT). Phosphatidylinositol glycan anchor biosynthesis class T (PIGT), a key GPIT subunit, influences GPI-APs biosynthesis and tumor biology. This study investigates PIGT expression in hepatocellular carcinoma (HCC) and its regulatory mechanisms.</p><p><strong>Methods: </strong>HCC genome sequencing and The Cancer Genome Atlas (TCGA) database were analyzed to compare PIGT expression between tumor and adjacent normal tissues. PIGT knockdown and overexpression cell lines examined its influence on HCC cell proliferation, migration, and invasion. Gene Set Enrichment Analysis (GSEA) identified downstream pathways, and Japan Australia Singapore Profiling Array Repository (JASPAR) predicted upstream transcriptional regulators, which were validated by in vivo tumor models.</p><p><strong>Results: </strong>PIGT was upregulated in HCC, enhancing tumor cell aggressiveness. GSEA implicated oncogenic pathways, and JASPAR identified homeobox B7 (HOXB7) as key transcriptional regulator. Animal models validated HOXB7-induced PIGT upregulation and its role in HCC progression.</p><p><strong>Conclusions: </strong>PIGT promotes HCC malignancy via the Wnt/β-catenin pathway, with HOXB7 as its upstream regulator.</p>","PeriodicalId":12099,"journal":{"name":"Expert Review of Anticancer Therapy","volume":" ","pages":""},"PeriodicalIF":2.8,"publicationDate":"2025-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144845056","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}
Sara Coca Membribes, Elizabeth Nally, Francesca Jackson-Spence, Catherine Graham, Salina Lalwani, Bernadett Szabados, Thomas Powles
{"title":"Established and emerging biomarkers approaches in urothelial carcinoma.","authors":"Sara Coca Membribes, Elizabeth Nally, Francesca Jackson-Spence, Catherine Graham, Salina Lalwani, Bernadett Szabados, Thomas Powles","doi":"10.1080/14737140.2025.2543454","DOIUrl":"10.1080/14737140.2025.2543454","url":null,"abstract":"<p><strong>Introduction: </strong>Urothelial carcinoma (UC) is marked by significant molecular heterogeneity and this complexity challenges precision medicine. Recent advances have improved biomarker development for UC diagnosis, prognosis and treatment.</p><p><strong>Areas covered: </strong>This review discusses established and emerging biomarkers in UC, including FGFR3 and HER2 alterations, PD-L1 expression and circulating tumor DNA (ctDNA). It also summarizes novel biomarkers such as Nectin-4, TROP-2, HER3, tumor mutational burden (TMB), and interferon-gamma signatures. The expanding role of artificial intelligence in biomarker discovery and interpretation is also addressed. Current literature was reviewed by a systematic search using PubMed, focusing on high-impact clinical trials, guidelines and recent reviews published up to May 2025.</p><p><strong>Expert opinion: </strong>Despite advances, clinical implementation of biomarkers in UC is limited by methodological inconsistencies and lack of standardization. Robust clinical trials and multi-modal approaches, including liquid biopsy, tissue analysis, and AI-driven tools, will be essential to advance precision oncology in UC.</p>","PeriodicalId":12099,"journal":{"name":"Expert Review of Anticancer Therapy","volume":" ","pages":"1-7"},"PeriodicalIF":2.8,"publicationDate":"2025-08-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144783874","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":"Prognostic significance of APOBEC1 and its role in lung squamous cell carcinoma: insights from chromatin regulator-based modeling and experimental validation.","authors":"Min Liang, Chaohao Zhang, Shengming Liu","doi":"10.1080/14737140.2025.2544869","DOIUrl":"10.1080/14737140.2025.2544869","url":null,"abstract":"<p><strong>Introduction: </strong>Chromatin regulators (CRs) are critical in cancer development, yet their prognostic value in lung squamous cell carcinoma (LUSC) remains unclear. This study aimed to develop a CR-based prognostic model and explore the role of APOBEC1 in LUSC progression.</p><p><strong>Methods: </strong>Transcriptomic and clinical data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) were analyzed. Gene Ontology and KEGG pathway enrichment were performed. Prognostic CRs were identified using univariate Cox and Lasso analyses. Drug sensitivity was assessed via the Drug Signatures Database. Key CRs were validated by PCR. APOBEC1 expression and prognosis were evaluated through pan-cancer analysis and experimentally validated in LUSC cell lines using colony formation, CCK-8, wound healing, and transwell assays.</p><p><strong>Results: </strong>An eight-gene CR-based signature was established, achieving 5-year AUCs of 0.87, 0.92, and 0.73 in the TCGA training, validation, and GEO datasets, respectively. High-risk patients showed enrichment in cancer-related pathways, greater immune infiltration, and elevated immune checkpoint expression. They were also more sensitive to Dasatinib, Bexarotene, and Bicalutamide. APOBEC1 was overexpressed across multiple cancer types and promoted proliferation and migration in LUSC cell lines.</p><p><strong>Conclusions: </strong>This study presents a robust CR-based survival model and highlights APOBEC1 as a potential therapeutic target in LUSC.</p>","PeriodicalId":12099,"journal":{"name":"Expert Review of Anticancer Therapy","volume":" ","pages":"1-14"},"PeriodicalIF":2.8,"publicationDate":"2025-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144783875","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}
Amer M Zeidan, Ruizhi Zhao, Robert S Epstein, Tehseen Salimi
{"title":"Oral decitabine and cedazuridine for the treatment of myelodysplastic syndromes: an integrated review of clinical, economic and patient-centered evidence.","authors":"Amer M Zeidan, Ruizhi Zhao, Robert S Epstein, Tehseen Salimi","doi":"10.1080/14737140.2025.2544859","DOIUrl":"10.1080/14737140.2025.2544859","url":null,"abstract":"<p><strong>Introduction: </strong>Myelodysplastic syndromes/neoplasms (MDS) are a heterogenous group of myeloid cancers that impose a substantial negative impact on patient health-related quality of life. As MDS predominately affects older individuals, who are especially susceptible to the debilitating nature of the disease and its burdensome symptoms, treatment decisions should consider therapeutic value from multiple perspectives to balance clinical efficacy with patient-centered outcomes. This comprehensive approach is known as relative medical value.</p><p><strong>Areas covered: </strong>Although improved outcomes have been observed with hypomethylating agents (HMAs) in patients with higher-risk MDS, parenteral administration of HMA requires frequent infusion clinic visits and is associated with substantial time burden, especially in older patients, impacting treatment persistence. Suboptimal use of HMAs in MDS may lead to poorer outcomes and higher healthcare costs, underscoring the need for patient-centered treatment options that improve persistence.</p><p><strong>Expert opinion: </strong>Oral decitabine and cedazuridine (DEC-C) is an approved treatment for higher-risk MDS and may reduce patient burden associated with parenteral HMA therapy. Oral DEC-C has the potential to improve treatment persistence and clinical outcomes and reduce healthcare resource utilization and costs. This review integrates the available clinical, patient-centered, and economic evidence on the relative medical value of oral DEC-C treatment for MDS.</p>","PeriodicalId":12099,"journal":{"name":"Expert Review of Anticancer Therapy","volume":" ","pages":"1-8"},"PeriodicalIF":2.8,"publicationDate":"2025-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144793856","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":"Epidemiologic and survival analysis for patients with secondary esophageal Cancer: a population-based study and external validation.","authors":"Jie Wang, Jingyun Zha, Xiaoliang Dong, Ping Liu","doi":"10.1080/14737140.2025.2517883","DOIUrl":"10.1080/14737140.2025.2517883","url":null,"abstract":"<p><strong>Background: </strong>There is a lack of dedicated studies addressing the epidemiology, prognostic factors, and optimal management strategies for secondary esophageal cancer (SEC).</p><p><strong>Methods: </strong>This study sourced data from the SEER database and an independent external cohort. Annual percentage change (APC) was calculated to qualify the incidence trend. Cox regression was employed for survival analysis.</p><p><strong>Results: </strong>A total of 13,792 sec cases from the SEER database and 63 cases from the validation cohort were included in this study. The incidence of SEC increased from 0.95 per 100,000 persons in 2000 to 1.2 per 100,000 in 2008, stabilizing thereafter. Survival analysis identified disease stage, age, and tumor location as significant prognostic factors (All <i>p</i> < .05). A prognostic model incorporating these variables demonstrated robust predictive accuracy across training, testing, and validation cohorts, with a C-index of around 0.73. In terms of treatment, radiotherapy and chemotherapy were associated with improved survival, while no significant benefit for younger patients and those with early-stage or squamous cell carcinoma.</p><p><strong>Conclusions: </strong>There remains a critical need for early detection methods and the implementation of more personalized treatment strategies. The developed prognostic model provides a framework for risk stratification, supporting the optimization of therapeutic decisions and improving patient outcomes.</p>","PeriodicalId":12099,"journal":{"name":"Expert Review of Anticancer Therapy","volume":" ","pages":"949-959"},"PeriodicalIF":2.8,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144247097","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}
Mei Chan, Tharani Krishnan, Amanda Townsend, Christos Karapetis, Amitesh Roy, Tiffany Foo, Niall Tebbutt, Tony Dhillon, Timothy Price
{"title":"The current landscape of neoadjuvant therapy for resectable colon cancer.","authors":"Mei Chan, Tharani Krishnan, Amanda Townsend, Christos Karapetis, Amitesh Roy, Tiffany Foo, Niall Tebbutt, Tony Dhillon, Timothy Price","doi":"10.1080/14737140.2025.2517881","DOIUrl":"10.1080/14737140.2025.2517881","url":null,"abstract":"<p><strong>Introduction: </strong>Neoadjuvant therapy has been proposed as a safe and effective treatment option for resectable colon cancer, providing several advantages over the current standard adjuvant protocol.</p><p><strong>Areas covered: </strong>This review summarizes the recent developments in neoadjuvant strategies for resectable colon cancer, highlighting key clinical trial data, current and emerging challenges, and the role of precision medicine in guiding treatment. Sources for this review were obtained through searches of PubMed, ClinicalTrials.gov, and relevant conference abstracts.</p><p><strong>Expert opinion: </strong>Neoadjuvant therapy is emerging as a promising approach for treating colon cancer, especially for dMMR/MSI-H colon cancer where neoadjuvant immunotherapy have shown exceptional complete pathologic response rates and durable responses. Ongoing trials are focused on identifying optimal treatment approaches that balance oncological efficacy with the minimization of toxicity. A reliable multimodal evaluation framework incorporating advanced staging techniques and biomarkers such as circulating tumor DNA is essential to guide treatment and improve patient selection.</p>","PeriodicalId":12099,"journal":{"name":"Expert Review of Anticancer Therapy","volume":" ","pages":"901-913"},"PeriodicalIF":2.8,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144247099","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":"An update on the therapeutic options for metastatic thymomas and thymic carcinomas.","authors":"Yoshihiro Masui, Yusuke Okuma","doi":"10.1080/14737140.2025.2519858","DOIUrl":"10.1080/14737140.2025.2519858","url":null,"abstract":"<p><strong>Introduction: </strong>Thymic epithelial tumors are rare cancers originating from the thymus, with an annual incidence of 0.13 per 100,000 persons. Therefore, the standard of care was approved primarily based on the results of single-arm phase II trials. If the disease is resectable and localized, a multidisciplinary treatment combining surgical resection, radiotherapy, and pharmacotherapy should be considered. Pharmacotherapy is the mainstay of treatment for metastatic and recurrent diseases.</p><p><strong>Areas covered: </strong>This review delineates the distinct difference in treatment strategies of chemotherapy in patients with thymomas and thymic carcinomas. Thymomas typically respond to a cisplatin and anthracyclines, often supplemented with steroids, whereas thymic carcinomas, which do not typically involve anthracyclines as key drugs, are treated with platinum-based doublet chemotherapy. Lenvatinib has emerged as a pivotal key drug for refractory thymic carcinoma. In addition, single-agent cytotoxic chemotherapy, molecular targeted therapies, and immune checkpoint inhibitors is considered as key drugs for thymic carcinomas.</p><p><strong>Expert opinion: </strong>Current research is focused on developing novel therapeutics such as antibody-drug conjugates (ADCs), angiogenesis inhibitors, multi-kinase inhibitors, and further immune checkpoint inhibitors, expanding the prospects for pharmacotherapy in thymic malignancies.</p>","PeriodicalId":12099,"journal":{"name":"Expert Review of Anticancer Therapy","volume":" ","pages":"853-863"},"PeriodicalIF":2.8,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144274567","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":"RASGEF1C affects aerobic glycolysis and facilitates lung cancer progression through the PLK1 signaling pathway.","authors":"Hao Wu, Kang Zheng, Fei Han","doi":"10.1080/14737140.2025.2518278","DOIUrl":"10.1080/14737140.2025.2518278","url":null,"abstract":"<p><strong>Objective: </strong>RasGEF domain family member 1C (RASGEF1C), primarily acting in neurological disorders, remains largely enigmatic regarding its function in lung cancer (LC).</p><p><strong>Methods: </strong>Bioinformatics analysis assessed the differential expression and prognosis of RASGEF1C in LC and analyzed the enriched pathways. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot (WB) analyses were employed to assess the expression of RASGEF1C and polo-like kinase 1 (PLK1). Biological functions were detected by cell counting kit-8 (CCK-8), flow cytometry, colony formation assay, cell scratch assay, and Transwell assay. Glycolysis-related proteins were detected by Western blot, and ATP levels, glucose content, lactate production, extracellular acidification rate, and oxygen consumption rate were measured to assess glycolysis flux changes in a mouse xenograft tumor model. Immunohistochemistry was applied to detect levels of RASGEF1C, PLK1, and Ki67. WB was employed to assess the expression of RASGEF1C, PLK1, pyruvate kinase M2 (PKM2), and hexokinase 2 (HK2).</p><p><strong>Results: </strong>The upregulation of RASGEF1C in LC reinforced the malignant progression of tumors (<i>p</i> < 0.01). RASGEF1C activation of the PLK1 pathway enhanced aerobic glycolysis, promoting proliferation, migration, and anti-apoptotic behaviors in LC cells.</p><p><strong>Conclusions: </strong>RASGEF1C affects aerobic glycolysis through the PLK1 signaling pathway, thereby acting as a pro-cancer factor driving LC progression.</p>","PeriodicalId":12099,"journal":{"name":"Expert Review of Anticancer Therapy","volume":" ","pages":"961-971"},"PeriodicalIF":2.8,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144368726","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}