Sandip M Prasad, Michael J Louie, Brent Burger, Victoria Tsurutis, Nikky Ugwuoke, Dalit Strauss-Ayali
{"title":"Pharmacokinetics of UGN‑102, an investigational mitomycin‑containing reverse thermal gel for the treatment of non-muscle invasive bladder cancer.","authors":"Sandip M Prasad, Michael J Louie, Brent Burger, Victoria Tsurutis, Nikky Ugwuoke, Dalit Strauss-Ayali","doi":"10.1007/s00280-025-04821-5","DOIUrl":"https://doi.org/10.1007/s00280-025-04821-5","url":null,"abstract":"<p><strong>Purpose: </strong>To evaluate the pharmacokinetic properties of UGN-102, a mitomycin-containing reverse thermal gel.</p><p><strong>Methods: </strong>Twelve patients with NMIBC received 6 once-weekly intravesical instillations of UGN-102 (BL004: 120 mg mitomycin [n = 6]; BL005: 75 mg mitomycin [n = 6]). Plasma samples for determination of pharmacokinetic (PK) parameters were collected up to 6 h following instillation.</p><p><strong>Results: </strong>In BL004, mean C<sub>max</sub> was 20.39 ng/mL, and median was 16.10 ng/mL (range 4.00-40.40), 100-10-fold lower than reported myelosuppression toxic level (RMTL); and median T<sub>max</sub> was 1.5 h. Mean AUC<sub>0-6</sub> was 56.23 ng·h/mL and mean apparent terminal half-life (t<sub>1/2</sub>) was 49 min. The highest observed C<sub>max</sub> (40.40 ng/mL) was 59-fold and 13-fold lower than C<sub>max</sub> following IV 30 mg or 10 mg mitomycin, respectively, and 10-fold lower than the RMTL. Maximum measured urine concentration was 635 µg/mL. In BL005, mean C<sub>max</sub> was 2.27 ng/mL, 181-fold lower than the RMTL; mean T<sub>max</sub> was 1.95 h and mean AUC<sub>0-6</sub> was 5.69 ng·h/mL. At 4-6 h post intravesical instillation, mitomycin concentrations in urine were either below or approaching the lower level of quantification (< 0.250 µg/mL) in 5/6 patients. In the remaining patient, the measurable urine concentrations over the 6-hour period suggest dwell time was > 6 h post-instillation, possibly due to the tumor location restricting urine flow. However systemic exposure was < 4 ng/mL, 100-fold lower than the RMTL.</p><p><strong>Conclusions: </strong>Intravesical instillation of UGN-102 results in low-level systemic absorption of mitomycin, with C<sub>max</sub> values considerably lower than those following IV administration and those associated with myelosuppression.</p><p><strong>Study registration: </strong>BL004: NCT02307487; BL005: NCT03558503.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":"95 1","pages":"100"},"PeriodicalIF":2.3,"publicationDate":"2025-10-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145273847","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jayesh Desai, Sanjeev Deva, Bo Gao, Kunyu Yang, Kenneth J O'Byrne, Meili Sun, Tianshu Liu, Tarek Meniawy, Xinmin Yu, Mark Voskoboynik, Diwakar Davar, Marco Matos, Shiangjiin Leaw, Tahmina Rahman, Xiaofei Qu, Hugh Giovinazzo, Xin Chen, Yan Dong, Daphne Day
{"title":"A phase I study of the OX40 agonist BGB-A445 with or without tislelizumab, an anti-PD-1 monoclonal antibody, in patients with advanced solid tumors: dose-escalation results.","authors":"Jayesh Desai, Sanjeev Deva, Bo Gao, Kunyu Yang, Kenneth J O'Byrne, Meili Sun, Tianshu Liu, Tarek Meniawy, Xinmin Yu, Mark Voskoboynik, Diwakar Davar, Marco Matos, Shiangjiin Leaw, Tahmina Rahman, Xiaofei Qu, Hugh Giovinazzo, Xin Chen, Yan Dong, Daphne Day","doi":"10.1007/s00280-025-04809-1","DOIUrl":"10.1007/s00280-025-04809-1","url":null,"abstract":"<p><strong>Purpose: </strong>OX40 may stimulate T-cell activation, potentially enhanced with checkpointinhibition. Results are from the dose-escalation part of an ongoing, multicenter, open-label study (NCT04215978, registered 30 December 2019) investigating OX40 agonist BGB-A445 alone or with anti-PD-1 antibody tislelizumab in patients with advanced solid tumors.</p><p><strong>Methods: </strong>Adults ≥18 years with previously treated advanced solid tumors, measurable by RECIST v1.1, enrolled. Dose-escalation A: 8 cohorts received increasing doses of IV BGB-A445 as monotherapy (20, 60, 150, 300, 600, 1200, 2400, 3600 mg); dose-escalation B: 6 cohorts received increasing doses of IV BGB-A445 (150, 300, 600, 1200, 2400, 3600 mg) + IV tislelizumab 200 mg. Primary objectives were safety and tolerability; secondary objectives included overall response rate (ORR) and pharmacokinetics.</p><p><strong>Results: </strong>112 patients were treated (A, n=63; B, n=49); 34/112 (30.4%) previously received checkpoint inhibitors. Treatment-related adverse events occurred in 36 (57.1%) (A) and 37 (75.5%) (B) patients, were grade ≥3 in 4 (6.3%) and 9 (18.4%), and caused treatment discontinuations in 1 (1.6%) and 1 (2.0%), respectively. Immune-mediated adverse events occurred in 8 (12.7%) (A) and 18 (36.7%) (B) patients, and infusion-related reactions in 9 (14.3%) (A) and 9 (18.4%) (B). No dose-limiting toxicities occurred. Unconfirmed ORR was 3.3% (unconfirmed partial response [PR], n=2) (A); confirmed was 21.3% (including PR, n=10) (B). BGB-A445 exposure increased dose-proportionally with a half-life of 8-13 days. OX40 receptor occupancy was saturated at ≥300 mg BGB-A445 in all cohorts.</p><p><strong>Conclusion: </strong>BGB-A445 was well tolerated and demonstrated on-target immune activation at clinically relevant doses. Antitumor activity was observed across cohorts.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":"95 1","pages":"99"},"PeriodicalIF":2.3,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12507935/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145249981","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Joshua Convert, Pierre Leblond, Perrine Marec-Berard, Anthony Ceraulo, Benoit Dumont, Michael Philippe
{"title":"Dosing chemotherapy in children with obesity: literature review and evaluation of local practices.","authors":"Joshua Convert, Pierre Leblond, Perrine Marec-Berard, Anthony Ceraulo, Benoit Dumont, Michael Philippe","doi":"10.1007/s00280-025-04825-1","DOIUrl":"https://doi.org/10.1007/s00280-025-04825-1","url":null,"abstract":"<p><strong>Purpose: </strong>Pediatric obesity is a growing public health concern, affecting drug pharmacokinetics, including chemotherapies. These challenges are amplified by the underrepresentation of children with obesity in clinical trials and the absence of clear dosing guidelines.</p><p><strong>Methods: </strong>A retrospective, single-center cohort study was conducted at the Institute of Pediatric Hematology and Oncology (Lyon, France), including children with obesity (Body mass index > IOTF-30) treated with at least one intravenous chemotherapy between 2008 and 2022. Medical records were reviewed for patient characteristics, chemotherapy type, administered dose and adjustments. A literature review was performed using PubMed to evaluate chemotherapy dosing recommendations for children with obesity.</p><p><strong>Results: </strong>A total of 248 chemotherapy courses for 113 children were analyzed, including 239 cytotoxic agents and 9 monoclonal antibodies. Most treatments were for hematological malignancies (n = 151), followed by solid tumors (n = 97). Only 45 chemotherapies (18%) were dose-adjusted in 19 patients (17%). Among these, 87% were capped at a body surface area (BSA) of 2 m², while 13% involved percentage-based reductions. Five adjustments were based on ideal body weight (IBW). Among the 25 patients with BSA > 2 m², 68% received adjusted doses, primarily through capping. In contrast, for patients with BSA < 2 m², dose reductions were rare and applied to avoid potential toxicity. The literature review revealed that total body weight (TBW) is generally used for dosing most chemotherapeutic agents, though certain drugs may require ideal body weight or adjusted body weight.</p><p><strong>Conclusion: </strong>Heterogeneity in clinical practice reflects the absence of standardized pediatric guidelines. Therapeutic drug monitoring and further pharmacokinetic studies may further help optimize dosing. TBW is recommended based on the study results and literature review, with exceptions, and guidelines should be widely disseminated to improve clinical practice.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":"95 1","pages":"98"},"PeriodicalIF":2.3,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145231654","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Modulation of prostaglandin-endoperoxide synthase-2 (PTGS2) mediated VEGF-signalling pathway by oral metronomic chemotherapy in locally advanced oral squamous cell Carcinoma: A brief report.","authors":"Mehta Vedant Kamal, Akhil Palod, Preetiparna Parida, Ananth Pai, Krishna Sharan, Vijetha Shenoy Belle, Rama Rao Damerla, Mahadev Rao, Naveena A N Kumar","doi":"10.1007/s00280-025-04819-z","DOIUrl":"https://doi.org/10.1007/s00280-025-04819-z","url":null,"abstract":"<p><strong>Background: </strong>Locally advanced oral squamous cell carcinoma (OSCC) is associated with poor survival outcomes, particularly in resource-limited settings, where treatment delays are prevalent. Oral metronomic chemotherapy (OMCT) utilising methotrexate and celecoxib is a potentially low-toxicity, cost-effective alternative in the neoadjuvant setting.</p><p><strong>Methods: </strong>Thirteen patients diagnosed with stage 3 or 4 resectable OSCC were enrolled in this study. Each participant received one month of OMCT prior to surgical intervention and continued OMCT as maintenance therapy following treatment. The expression levels of PTGS2, VEGFA, VEGFB, KDR, CXCR1, and CXCR2 were analysed both before and after OMCT administration. Additionally, organ function and patient survival were evaluated to determine the efficacy and toxicity of treatment.</p><p><strong>Results: </strong>PTGS2, VEGFA, VEGFB, and KDR were significantly upregulated in the tumour tissues at baseline. Post-OMCT, VEGFA, VEGFB, and KDR were significantly downregulated, whereas PTGS2 expression increased. No significant changes were observed in CXCR1 and CXCR2 expression. Liver, renal, and thyroid functions remained within normal limits. No adverse events were reported. The median overall survival was 22.1 months, and the median disease-free survival was 20 months. Upregulation of PTGS2, VEGFA, and VEGFB was correlated with improved outcomes.</p><p><strong>Conclusion: </strong>OMCT has the potential to manage disease progression in patients with OSCC awaiting surgical intervention. It is characterised by low toxicity and may exert anti-angiogenic effects through modulation of the VEGF pathway. Further large-scale trials are necessary to substantiate these findings and to establish optimal treatment strategies.</p><p><strong>Trial registration: </strong>The study was also registered in the Clinical Trials Registry of India (registration number: CTRI/2021/01/030256).</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":"95 1","pages":"96"},"PeriodicalIF":2.3,"publicationDate":"2025-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145225254","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Małgorzata Pieniążek, Marcin Kubeczko, Manuela Las-Jankowska, Anna Polakiewicz-Gilowska, Aleksandra Łacko, Michał Jarząb, Zuzana Bielčiková, Renata Pacholczak-Madej, Miroslawa Püsküllüoğlu
{"title":"Sacituzumab Govitecan initial dose reduction in polish patients with metastatic triple-negative breast cancer: impact on efficacy and safety.","authors":"Małgorzata Pieniążek, Marcin Kubeczko, Manuela Las-Jankowska, Anna Polakiewicz-Gilowska, Aleksandra Łacko, Michał Jarząb, Zuzana Bielčiková, Renata Pacholczak-Madej, Miroslawa Püsküllüoğlu","doi":"10.1007/s00280-025-04823-3","DOIUrl":"10.1007/s00280-025-04823-3","url":null,"abstract":"<p><strong>Background: </strong>Sacituzumab govitecan (SG) is approved for metastatic triple-negative breast cancer in ≥ 2 line setting at 10 mg/kg IV on Days 1 and 8 (21-day cycle). Trials confirmed its superiority over 8 mg/kg with manageable safety. In practice, precautionary dose reductions are used despite no formal guidance. In Poland, fixed 200 mg vials and unreimbursed drug waste lead to early dose adjustments.</p><p><strong>Methods: </strong>This retrospective study evaluated the impact of initial SG dose reduction on treatment outcomes and tolerability in Polish patients. Medical records provided data on baseline features, treatment, survival, and safety. Kaplan-Meier and chi-square tests were used for survival and group comparisons. A multivariate Cox model assessed the independent effect of dose reduction on overall survival (OS) and progression-free survival (PFS). Significance was set at p < 0.05.</p><p><strong>Results: </strong>Among 83 patients (median age 55, range 30-86), initial dose reductions ≥ 10% were observed in 16 patients (19.3%), including 9 (10.8%) with dose reduced ≥ 20%. Administrative adjustments (reductions > 10% to flat doses of 200 mg multiplications) accounted for 18.1% of the entire cohort. Grade ≥ 2 and ≥ 3 adverse events occurred in 83.1% and 56.6%, respectively. In a multivariate analysis, a ≥ 20% initial dose reduction remained an independent predictor of shorter PFS (HR: 2.6; 95% CI: 1.1-6.6; p = 0.04) and OS (HR: 6; 95% CI: 2-17.5; p = 0.001). Initial dose reduction did not affect toxicity.</p><p><strong>Conclusions: </strong>In this preliminary report initial dose reduction of SG negatively impacted PFS and OS without reducing toxicity, highlighting the need for further studies and dosing policy adjustments.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":"95 1","pages":"97"},"PeriodicalIF":2.3,"publicationDate":"2025-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12496269/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145228275","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Comment on: Dose adjustment strategy for high dose methotrexate-induced toxicities in pediatric acute lymphoblastic leukemia: based on population PK analysis and exposure toxicity relationship.","authors":"Ian J Cohen","doi":"10.1007/s00280-025-04813-5","DOIUrl":"https://doi.org/10.1007/s00280-025-04813-5","url":null,"abstract":"","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":"95 1","pages":"95"},"PeriodicalIF":2.3,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145211935","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Effects of infliximab, an anti-TNF-α, on cyclophosphamide-induced bone marrow toxicity.","authors":"Koorosh Tabatabaei, Haleh Vaez, Amirhosein Ghafouri-Asbagh, Bahram Amouoghli Tabrizi, Mir Alireza Nourazar, Behrooz Shokouhi","doi":"10.1007/s00280-025-04814-4","DOIUrl":"https://doi.org/10.1007/s00280-025-04814-4","url":null,"abstract":"<p><strong>Purpose: </strong>Cyclophosphamide is a widely used anticancer agent, though its clinical application is often constrained by myelotoxicity. Infliximab, a monoclonal antibody targeting TNF-α, may alleviate this toxicity by mitigating oxidative stress. This study aimed to assess the protective effects of infliximab against cyclophosphamide-induced myelotoxicity, focusing on bone marrow and spleen-related parameters.</p><p><strong>Methods: </strong>Male Wistar rats (260 ± 20 g) were randomly divided into four groups (n = 5 each): Control (normal saline), Infliximab (7 mg/kg, IP), Cyclophosphamide (200 mg/kg, IP), and Infliximab + Cyclophosphamide (same respective doses with a four-day interval between administrations). Nine days after the initiation of the experiment, bone marrow and spleen histopathology, bone marrow cellularity, spleen index, and levels of superoxide dismutase (SOD) and malondialdehyde (MDA) in bone marrow tissue, along with serum TNF-α and G-CSF concentrations, were evaluated.</p><p><strong>Results: </strong>Infliximab prevented myelofibrosis in bone marrow tissue; however, it did not mitigate cellularity depletion. It failed to counteract the effects of cyclophosphamide on spleen histology and the spleen index. Additionally, infliximab enhanced antioxidant activity in the bone marrow. Moreover, inhibition of TNF-α was associated with an increase in G-CSF levels.</p><p><strong>Conclusion: </strong>Although infliximab improved molecular pathways associated with cyclophosphamide-induced bone marrow and hematologic toxicity in the rat model, it did not lead to significant improvements in clinical manifestations.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":"95 1","pages":"94"},"PeriodicalIF":2.3,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145211884","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"High MCM10 expression mediates TMZ resistance and promotes the progression of glioma.","authors":"Qiusi Tian, Zhijun Bao, Yifei Zhao, Qun Zhang","doi":"10.1007/s00280-025-04807-3","DOIUrl":"https://doi.org/10.1007/s00280-025-04807-3","url":null,"abstract":"<p><strong>Background: </strong>In glioblastoma, the heterogeneity of tumors often complicates treatment outcomes, particularly resistance to temozolomide (TMZ). This study aimed to investigate the expression characteristics of MCM10 in gliomas and its potential role in TMZ resistance.</p><p><strong>Methods: </strong>Analyze the expression of MCM10 in glioma cells within the datasets Glioma_GSE102130, Glioma_GSE130224, Glioma_GSE131928_10X, and Glioma_GSE131928_Smartseq2 using the TISCH2 single-cell database.Subsequently, the expression level of MCM10 was evaluated using the TMZ-resistant cell lines U251-TR and LN229-TR. Glioma cells with reduced MCM10 expression were constructed through lentiviral interference.Construct glioma cells with MCM10 knockdown through lentiviral interference. Additionally, this study conducts an association analysis between MCM10 expression and patterns of DNA methylation and RNA methylation.Finally, the function of MCM10 was validated in a heterotopic mouse model.</p><p><strong>Results: </strong>MCM10 mRNA is highly expressed in osteoclast-like malignant cells (OC-likemalignant cells) and oligodendrocyte progenitor-like malignant cells (OPC-like malignant cells). After treatment with TMZ, the levels of MCM10 protein and mRNA significantly increased. Knocking down MCM10 significantly reduced the migration and invasion of U251 and LN229 cells, while altering the expression of molecular markers associated with epithelial-mesenchymal transition (EMT). Weighted gene co-expression network analysis (WGCNA) revealed signaling pathways associated with drug resistance and identified overlapping differentially expressed mRNAs between resistant cells and MCM10 knockdown cells. High expression of MCM10 is associated with low DNA methylation, and MCM10 methylation is positively correlated with patient survival rates. Furthermore, inhibition of MCM10 significantly slowed tumor growth, indicating its potential as a therapeutic target for gliomas.</p><p><strong>Conclusion: </strong>MCM 10 is a key mediator of TMZ resistance in glioblastoma.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":"95 1","pages":"93"},"PeriodicalIF":2.3,"publicationDate":"2025-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145198587","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Haruhiko Yamazaki, Nobuyasu Suganuma, Mei Kadoya, Katsuhiko Masudo, Soji Toda, Aya Saito
{"title":"Tumor response and thyroglobulin change in differentiated thyroid carcinoma treated with dabrafenib plus trametinib.","authors":"Haruhiko Yamazaki, Nobuyasu Suganuma, Mei Kadoya, Katsuhiko Masudo, Soji Toda, Aya Saito","doi":"10.1007/s00280-025-04824-2","DOIUrl":"https://doi.org/10.1007/s00280-025-04824-2","url":null,"abstract":"<p><strong>Purpose: </strong>It has been reported that treatment response does not necessarily correlate with the change of thyroglobulin (Tg) during dabrafenib treatment in differentiated thyroid carcinoma (DTC). This study aimed to assess the association between the clinical response and Tg changes or inflammatory biomarkers in a real-world setting.</p><p><strong>Methods: </strong>This retrospective multi center cohort study included 22 BRAF-mutated DTC patients treated with dabrafenib plus trametinib in three academic institutions.</p><p><strong>Results: </strong>All 22 patients harbored the BRAFV600E mutation. Twenty-one patients (95%) had papillary thyroid carcinoma histology, and one had poorly differentiated thyroid carcinoma histology. Among 16 patients without Tg antibody, 14 patients (88%) experienced an increase in their Tg levels one month after the initiation of dabrafenib plus trametinib. In addition, 11 patients (69%) experienced an increase in their Tg levels at the best clinical response. Among these 11 patients who had an increase in Tg level at the best clinical response, the numbers of patients who had partial response, stable disease, and progressive disease were 3, 8, and 0, respectively. Among the 19 patients in whom neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, and lymphocyte-to-monocyte ratio were measured at baseline, no distinctive trends were identified between clinical response and change in those inflammatory biomarkers.</p><p><strong>Conclusions: </strong>Tg changes during dabrafenib plus trametinib treatment may not be associated with clinical response to dabrafenib plus trametinib treatment. Further study is needed to clarify the association between Tg level or inflammatory biomarkers change and clinical response to dabrafenib plus trametinib treatment.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":"95 1","pages":"92"},"PeriodicalIF":2.3,"publicationDate":"2025-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145197788","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Non-oncologic to oncologic drug: A systematic review of drug repurposing in cancer.","authors":"Yarava Dhanush, Vakkalagadda Siva Ganesh","doi":"10.1007/s00280-025-04822-4","DOIUrl":"10.1007/s00280-025-04822-4","url":null,"abstract":"<p><strong>Purpose: </strong>This systematic review investigates drug repurposing as a strategy to accelerate and improve cancer treatment by specifically examining how existing medications can effectively target the established hallmarks of cancer. The research explores how repurposed drugs can address the challenges of conventional cancer drug development, including high costs, lengthy development timelines, and frequent clinical failures.</p><p><strong>Methods: </strong>The review systematically analyzes repurposed drugs with their ability to target specific cancer hallmarks, including oncogenic signaling pathways, cell death regulation, metabolic reprogramming, growth suppressor reactivation, phenotypic plasticity, antitumor immunity, telomerase activity, angiogenesis, inflammation, cellular senescence, invasion and metastasis, DNA damage response, microbiome modulation, and epigenetic regulation.</p><p><strong>Results: </strong>The analysis identified several promising repurposed medications targeting specific cancer hallmarks: artemisinin derivatives for oncogenic signalling, niclosamide for cell death pathways, leflunomide for metabolic dysregulation, statins for tumour suppressor reactivation, metformin for phenotypic plasticity, liothyronine for immune activation, PARP inhibitors for replication, itraconazole for angiogenesis, celecoxib for inflammation, BCL-2 inhibitors for senescence, fluoroquinolones for metastasis, spironolactone for DNA damage, isoliquiritigenin for microbiome modulation, and various agents targeting non-mutational epigenetic regulation.</p><p><strong>Conclusion: </strong>Drug repurposing represents an intriguing approach for addressing the limitations of traditional cancer drug development while effectively targeting the fundamental hallmarks of cancer. By leveraging existing medications with established safety profiles, this strategy offers potential for more rapid translation to clinical applications and may enhance the therapeutic arsenal against various cancer types.</p>","PeriodicalId":9556,"journal":{"name":"Cancer Chemotherapy and Pharmacology","volume":"95 1","pages":"91"},"PeriodicalIF":2.3,"publicationDate":"2025-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145173825","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}