Medical OncologyPub Date : 2025-09-23DOI: 10.1007/s12032-025-03056-x
Haohan Li, Yuan Wang, Xinghe Tong, Yanlong Yang, Yinsong Tian, Jie Jia, Tao He, Rui Liu, Xudong Yang, Xiaobo Chen
{"title":"The dual role of PCDH9 in tumors, neurological and developmental diseases.","authors":"Haohan Li, Yuan Wang, Xinghe Tong, Yanlong Yang, Yinsong Tian, Jie Jia, Tao He, Rui Liu, Xudong Yang, Xiaobo Chen","doi":"10.1007/s12032-025-03056-x","DOIUrl":"https://doi.org/10.1007/s12032-025-03056-x","url":null,"abstract":"<p><p>Protocadherin 9 (PCDH9), a member of the δ1-protocadherin family, plays a crucial role in regulating cell polarity, tumor suppression, and neurodevelopment. This is achieved through its dual functions of extracellular calcium-dependent adhesion and intracellular signal transduction. In the context of tumors, PCDH9 inhibits epithelial-mesenchymal transition and cell cycle progression in various cancer types, such as liver cancer and glioma, via the GSK-3β/Snail1 axis. However, in Group 4 medulloblastoma, functionally acquired mutations in PCDH9 drive cancer development through the non-classical Wnt pathway. The expression of PCDH9 is tightly regulated by microRNAs (e.g., miR-589-3p) and epigenetic silencing mechanisms. Within the nervous system, PCDH9 deficiency gives rise to abnormal autistic behaviors, disruptions in hippocampal migration, and abnormal cerebrospinal fluid circulation. Its potential for clinical translation is evident in several areas, including the use of methylation as a prognostic marker, β-eucalyptol-mediated therapy to restore PCDH9, and strategies that target the piRNA-PI3K/AKT axis. Looking ahead, it is essential to conduct in-depth analyses of its evolutionary adaptability and formulate strategies targeting the adherent-signal interface.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"489"},"PeriodicalIF":3.5,"publicationDate":"2025-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145124907","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}
Medical OncologyPub Date : 2025-09-23DOI: 10.1007/s12032-025-03051-2
Fahadul Islam, Mehrukh Zehravi, M Raju Molla, Sherouk Hussein Sweilam, Patibandla Jahnavi, Om Mahadeo Bagade, Jeetendra Kumar Gupta, Rajeshwar Vodeti, Karthickeyan Krishnan, Vasudha Bakshi, P Dharani Prasad, Muath Suliman, Mst Nowsad Zahan Sathi, Talha Bin Emran
{"title":"Stilbenes in cancer therapy: insights into molecular targets, and advances towards clinical application.","authors":"Fahadul Islam, Mehrukh Zehravi, M Raju Molla, Sherouk Hussein Sweilam, Patibandla Jahnavi, Om Mahadeo Bagade, Jeetendra Kumar Gupta, Rajeshwar Vodeti, Karthickeyan Krishnan, Vasudha Bakshi, P Dharani Prasad, Muath Suliman, Mst Nowsad Zahan Sathi, Talha Bin Emran","doi":"10.1007/s12032-025-03051-2","DOIUrl":"https://doi.org/10.1007/s12032-025-03051-2","url":null,"abstract":"<p><p>Stilbenes are polyphenolic substances that are found in plants such as peanuts, grapes, and berries. Some of these compounds have demonstrated strong anticancer potential via multiple mechanisms. Pterostilbene and resveratrol have been shown to inhibit cancer cell proliferation, promote apoptosis, and inhibit the spread of cancer to other parts of the body. They can disrupt important molecular pathways, which ultimately results in the stoppage of the cell cycle, the activation of apoptosis, the prevention of angiogenesis, and the suppression of tumor invasion and metastasis. Stilbenes also possess powerful anti-inflammatory and antioxidant potential, which leads to their anticancer effects by lowering chronic inflammation and oxidative stress. Stilbenes are associated with multiple health benefits. On the other hand, their quick metabolism and low bioavailability make it difficult to use them in clinical settings successfully. The solubility, stability, and targeted distribution of these substances have been improved via the development of synthetic derivatives and nanoformulations by researchers. Stilbenes and their derivatives are valuable as innovative chemopreventive and therapeutic agents in the field of cancer due to their ability to target several targets simultaneously and their low toxicity.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"487"},"PeriodicalIF":3.5,"publicationDate":"2025-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145124941","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}
Medical OncologyPub Date : 2025-09-20DOI: 10.1007/s12032-025-03005-8
Parteek Prasher, Mousmee Sharma, Rabab Fatima, William N Setzer, Javad Sharifi-Rad
{"title":"Bisabolol as a natural anticancer agent: molecular insights and therapeutic potential in oncology.","authors":"Parteek Prasher, Mousmee Sharma, Rabab Fatima, William N Setzer, Javad Sharifi-Rad","doi":"10.1007/s12032-025-03005-8","DOIUrl":"https://doi.org/10.1007/s12032-025-03005-8","url":null,"abstract":"<p><p>α-Bisabolol is a naturally derived monocyclic sesquiterpene, abundantly present in German chamomile (Matricaria recutita) and various other aromatic plants and is also increasingly accessible via metabolic engineering platforms. The physicochemical profile and ADMET characteristics of α-bisabolol, suggest a high gastrointestinal absorption, and minimal P-glycoprotein and CYP450 interactions, which validate its drug-like potential. The multi-protein target engagement and predicted activity spectrum of α-bisabolol have been discussed in the manuscript by using in silico tools (SwissTargetPrediction and PASS), which aligns with the experimental anticancer results of α-bisabolol in leukemia, pancreatic, lung, and glioblastoma models. Mechanistically, the anticancer profile of α-bisabolol arises from the induction of mitochondrial apoptosis, disruption of PI3K/Akt/FAK/BRAF pathways, modulation of lipid-raft-associated Bid protein, and dysregulation of autophagy. The design and bioactivity of novel chemical derivatives (e.g., acyl, glycoside, and thiosemicarbazone analogs) and delivery systems (cyclodextrin inclusion complexes, PLGA nanoparticles), emphasizing improvements in solubility, potency, and systemic delivery have also been comprehensively discussed in this review. Finally, a critical evaluation of the clinical translation barriers including poor aqueous solubility, limited in vivo pharmacokinetics, CYP2C9/CYP2J-mediated metabolism, and formulation challenges of α-bisabolol have been succinctly provided. Overall, this review integrates phytochemistry, polypharmacology, mechanistic validation, and translational hurdles, thereby offering a novel roadmap for developing α-bisabolol as a viable anticancer therapeutic.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"485"},"PeriodicalIF":3.5,"publicationDate":"2025-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145092065","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}
Medical OncologyPub Date : 2025-09-20DOI: 10.1007/s12032-025-03042-3
Ahmad M Alamri, Abdullah A Assiri, Bushra Khan, Najeeb Ullah Khan
{"title":"Next-generation oncology: integrative therapeutic frontiers at the crossroads of precision genomics, immuno-engineering, and tumor microenvironment modulation.","authors":"Ahmad M Alamri, Abdullah A Assiri, Bushra Khan, Najeeb Ullah Khan","doi":"10.1007/s12032-025-03042-3","DOIUrl":"https://doi.org/10.1007/s12032-025-03042-3","url":null,"abstract":"<p><p>The landscape of oncology is undergoing a paradigm shift, transitioning from conventional cytotoxic therapies to an integrative, intelligence-driven framework that combines precision genomics, immunoengineering, and modulation of the tumor microenvironment (TME). This review explores how cancer, as a complex adaptive system (CAS), evolves through genetic, epigenetic, and microenvironmental interactions, necessitating dynamic, multi-dimensional therapeutic strategies. Review highlights the limitations of mono-targeted therapies and the emergence of synergistic approaches, including AI-guided adaptive dosing, synthetic biology-enhanced CAR-T cells, and metabolic reprogramming of the tumor microenvironment (TME). Breakthroughs in molecular cartography, quantum biology, synthetic oncology, and dark genome mining are expanding therapeutic frontiers. Meanwhile, immuno-engineering innovations-such as next-generation checkpoint modulators, logic-gated CAR-T cells, and neoantigen vaccines-are redefining immune-oncology. Additionally, TME-targeted strategies, including stromal remodeling, hypoxia modulation, and microbiome engineering, are helping to overcome treatment resistance. The convergence of multi-omics profiling, combinatorial therapeutics, and computational oncology (e.g., digital twins) is enabling real-time, personalized interventions. Despite these advances, challenges persist-therapeutic resistance, toxicity, accessibility, and ethical concerns-demanding interdisciplinary collaboration and equitable innovation. The future lies in adaptive, autonomous oncology, integrating AI, closed-loop therapies, and modular mRNA platforms to deliver precision medicine at scale. This review underscores the imperative for a unified, systems-based approach to transform cancer into a manageable condition.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"482"},"PeriodicalIF":3.5,"publicationDate":"2025-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145092092","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}
Medical OncologyPub Date : 2025-09-20DOI: 10.1007/s12032-025-03037-0
Praveen Dhyani, Priyanka Sati, Dharam Chand Attri, Eshita Sharma, Erica Campagna, Maria Atanassova, Gianluca Caruso, Zainab M Almarhoon, Daniela Calina, William N Setzer, Javad Sharifi-Rad
{"title":"Apratoxin S10 as a dual-action modulator of receptor tyrosine kinases and tumor microenvironment: emerging anticancer insights from marine-derived analogs.","authors":"Praveen Dhyani, Priyanka Sati, Dharam Chand Attri, Eshita Sharma, Erica Campagna, Maria Atanassova, Gianluca Caruso, Zainab M Almarhoon, Daniela Calina, William N Setzer, Javad Sharifi-Rad","doi":"10.1007/s12032-025-03037-0","DOIUrl":"https://doi.org/10.1007/s12032-025-03037-0","url":null,"abstract":"<p><p>Marine cyanobacteria are prolific producers of structurally diverse and pharmacologically potent secondary metabolites. Among these, apratoxins, a class of cyclodepsipeptides originally isolated from Lyngbya species have demonstrated broad-spectrum cytotoxic and antiangiogenic activity. However, the clinical development of natural apratoxins has been limited due to systemic toxicity and narrow therapeutic indices. Recent efforts have focused on optimizing these molecules, leading to the development of semi-synthetic analogs such as Apratoxin S10 (Apra S10), which exhibits improved stability, selectivity, and potency. This review synthesizes current evidence on the anticancer mechanisms of Apra S10 and related apratoxins, including their effects on receptor tyrosine kinases (RTKs), growth factor signaling, and tumor microenvironment modulation. Emphasis is placed on Apra S10's preclinical pharmacokinetics, tumor-specific accumulation, and multi-target activity across highly vascularized tumors, including hepatocellular carcinoma, renal cell carcinoma, neuroendocrine tumors, and pancreatic ductal adenocarcinoma. Studies show that Apra S10 downregulates RTKs, suppresses secretion of VEGF-A and IL-6, and disrupts tumor-stroma crosstalk, mechanisms that collectively result in potent growth inhibition and antiangiogenic effects without overt toxicity. These findings highlight Apra S10 and its analogs as promising candidates for adjuvant cancer therapy, meriting further translational research to assess clinical safety, pharmacodynamics, and synergistic potential with existing chemotherapeutics.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"480"},"PeriodicalIF":3.5,"publicationDate":"2025-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145092075","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}
Medical OncologyPub Date : 2025-09-20DOI: 10.1007/s12032-025-03041-4
Ildar W Mutalip, Allyson F Barth, Garen S Sellers, Mark R Wakefield, Yujiang Fang
{"title":"From bench to bedside: the evolution and future of prostate-specific antigen testing.","authors":"Ildar W Mutalip, Allyson F Barth, Garen S Sellers, Mark R Wakefield, Yujiang Fang","doi":"10.1007/s12032-025-03041-4","DOIUrl":"https://doi.org/10.1007/s12032-025-03041-4","url":null,"abstract":"<p><p>Prostate cancer is the most common cancer in males in the U.S. except skin cancer. Prostate-Specific Antigen (PSA) is a glycoprotein enzyme produced by the prostate epithelial cells that has become a pivotal biomarker in the diagnosis, monitoring, and management of prostate cancer. Since its identification in seminal plasma in the early 1970s and subsequent purification from prostate, PSA has been widely adopted in clinical practice for screening for prostate cancer, particularly after FDA approval for this purpose in 1994. This study reviewed the historical development of PSA, its biochemical and physiological properties, and its clinical applications in cancer detection, staging, and monitoring. The advantages of PSA testing, including its role in early detection and reduction of prostate cancer mortality, are balanced against limitations such as false positives, overdiagnosis, and overtreatment. Recent innovations, including PSA derivatives, imaging techniques, urinary biomarkers, and risk calculators, are also discussed as tools to improve accuracy in diagnoses. As research progresses, PSA remains a valuable, though imperfect, component of prostate cancer care, with ongoing efforts aimed at enhancing its specificity and integrating it into personalized screening strategies. Such a study would be more helpful for clinicians to effectively use PSA as a screening biomarker and/or monitoring biomarker for prostate cancer.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"486"},"PeriodicalIF":3.5,"publicationDate":"2025-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145092073","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":"Kinesin superfamily proteins in ovarian cancer: from molecular mechanisms to clinical applications.","authors":"Ashok Kumar Bishoyi, Shaker Al-Hasnaawei, Subbulakshmi Ganesan, Aman Shankhyan, Anima Nanda, Aashna Sinha, Subhashree Ray, Deepak Nathiya","doi":"10.1007/s12032-025-03044-1","DOIUrl":"https://doi.org/10.1007/s12032-025-03044-1","url":null,"abstract":"<p><p>Ovarian cancer remains one of the most lethal malignancies affecting women, largely due to its asymptomatic onset and frequent diagnosis at advanced stages. Emerging evidence has underscored the pivotal role of kinesin superfamily proteins (KIFs) in orchestrating the cellular mechanisms underlying tumor initiation and progression, particularly in ovarian cancer. These microtubule-associated motor proteins are essential for intracellular transport, cell division, signal transduction, and organelle positioning. Dysregulation of KIFs has been implicated in enhanced cellular proliferation, metastasis, and resistance to chemotherapy. In ovarian cancer, specific KIF members have been shown to promote cell motility and interfere with key signaling pathways, thereby accelerating tumor progression. Elevated expression levels of certain KIFs correlate with poor patient prognosis and reduced overall survival. This review consolidates current insights into the role of KIFs in ovarian cancer pathogenesis and emphasizes their potential as therapeutic targets. Elucidating the mechanistic involvement of KIFs in this malignancy may pave the way for innovative diagnostic, prognostic, and therapeutic strategies aimed at improving clinical outcomes in this highly aggressive cancer type.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"483"},"PeriodicalIF":3.5,"publicationDate":"2025-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145092095","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}
Medical OncologyPub Date : 2025-09-20DOI: 10.1007/s12032-025-03040-5
Mohamed Saad Sayed, Ahmed Farid Gadelmawla, Osama Abouelenin, Elsayed S Moubarak, Nada S Jibril, Mahmoud Kandeel, Hebatullah Abdulazeem
{"title":"Chimeric antigen receptor T-cell (CAR T-cell) and tumor-infiltrating lymphocytes (TILs) therapies in gastrointestinal malignancies: review of literature for clinical applications.","authors":"Mohamed Saad Sayed, Ahmed Farid Gadelmawla, Osama Abouelenin, Elsayed S Moubarak, Nada S Jibril, Mahmoud Kandeel, Hebatullah Abdulazeem","doi":"10.1007/s12032-025-03040-5","DOIUrl":"10.1007/s12032-025-03040-5","url":null,"abstract":"<p><p>Gastrointestinal malignancies (GI malignancies) have had a notoriously dismal prognosis throughout history. The primary therapeutic approaches to treat and manage GI malignancies are immunotherapy, radiotherapy, surgery, and chemotherapy, which may include monotherapy or a combination of these therapies to boost the effect. Nevertheless, the recurrence and metastasis rates remain elevated. In recent decades, immunotherapies have had a powerful impact when included in treatment regimens. In hematologic malignancy, chimeric antigen receptor T cells (CAR-T cell) have shown a promising anticancer impact as one of the immunotherapies. It gives a promising treatment option for solid tumors, including colorectal cancers. In recent clinical trials, the CAR-T cells showed a promising effect on pancreatic, colorectal, esophageal, hepatocellular, and gastric cancers. Tumor-infiltrating lymphocyte (TIL) therapy is another immunotherapy option with promising option for GI malignancies. Through the process of designing the TIL therapy, T cells are extracted and designed according to the nature of the GI malignancy. In this review, we addressed the clinical applications of both therapies while highlighting the challenges and possible strategies to overcome them. CAR T-cells and TIL therapies showed good responses with tolerable and acceptable side effects in treating GI malignancies such as pancreatic, colorectal, gastric, and hepatocellular cancers, while the immunosuppressive tumor microenvironment (TME) inhibiting the activity of immunotherapy and impeding its efficacy is a significant challenge.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"481"},"PeriodicalIF":3.5,"publicationDate":"2025-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12450226/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145092098","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}
Medical OncologyPub Date : 2025-09-20DOI: 10.1007/s12032-025-03048-x
Huiping Li, Yangli Jin, Qing Huang, Xiaohua Xie, Nan Li
{"title":"Comprehensive analysis of VDAC1 in gynecological tumors and structure-based virtual screening of its natural inhibitors.","authors":"Huiping Li, Yangli Jin, Qing Huang, Xiaohua Xie, Nan Li","doi":"10.1007/s12032-025-03048-x","DOIUrl":"https://doi.org/10.1007/s12032-025-03048-x","url":null,"abstract":"<p><p>The voltage-dependent anion channel 1 (VDAC1) protein is an important regulator of mitochondrial function. The aim of this study was to investigate the role of VDAC1 in gynecological malignancies, and to screen the natural compounds targeting VDAC1, as the candidate anti-cancer drugs. The expression levels of VDAC1 mRNA and protein were analyzed using the GEPIA and UALCAN databases. The TISIDB database was used to analyze the correlation between VDAC1 expression and tumor stage, histological grade and immunomodulators. The pan-cancer prognostic value of VDAC1 was evaluated using the GEPIA2 database. Genetic Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed using the R software clusterProfiler package. The relationship between VDAC1 expression and immune cell infiltration in gynecological tumors was analyzed using the TIMER database. Potential compounds targeting VDAC1 were screened by virtual screening and molecular docking. After VDAC1 was silenced or aloe-emodin treatment, the expressions of VDAC1, Bcl-2 and Bax in cancer cells were detected by RT-qPCR and Western blot, and cancer cell proliferation was detected by CCK-8 assay, and apoptosis was evaluated by flow cytometry, and migration of the cancer cells were examined with Transwell assay. It was revealed that, VDAC1 was highly expressed in cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC), uterine corpus endometrial carcinoma (UCEC) and ovarian cancer (OV). High expression of VDAC1 was associated with higher tumor stage and poor prognosis of CESC, as well as high histological grade and immune subtypes of UCEC. Knocking down VDAC1 repressed the proliferation and migration of CESC, UCEC and OV cells, and promoted the apoptosis of tumor cells. Aloe-emodin showed strong binding affinity with VDAC1 protein, and it showed tumor-suppressive properties against CESC, UCEC and OV cells. In conclusion, VDAC1 may be a potential diagnostic biomarker and a new target for gynecological malignancies, and aloe-emodin is a candidate anti-cancer drug targeting VDAC1.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"484"},"PeriodicalIF":3.5,"publicationDate":"2025-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145092103","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}
Medical OncologyPub Date : 2025-09-19DOI: 10.1007/s12032-025-03054-z
Daniel Muteb Muyey, Yuanyuan Wang, Shuo Li, Zhuanghui Hao, Jingyi Feng, Fanggang Ren, Hongwei Wang
{"title":"Synergy Between second-generation FLT3 inhibitors and the ERK1/2 inhibitor Ulixertinib in FLT3-ITD-mutated acute myeloid leukemia (AML) cells.","authors":"Daniel Muteb Muyey, Yuanyuan Wang, Shuo Li, Zhuanghui Hao, Jingyi Feng, Fanggang Ren, Hongwei Wang","doi":"10.1007/s12032-025-03054-z","DOIUrl":"https://doi.org/10.1007/s12032-025-03054-z","url":null,"abstract":"<p><p>The FLT3 internal tandem duplication (FLT3-ITD) mutation is a critical molecular marker in acute myeloid leukemia (AML) and is closely associated with adverse patient prognosis. Although FLT3 inhibitors have been clinically applied, their therapeutic efficacy is constrained by reduced drug responsiveness and disease relapse. This study aims to investigate the underlying causes of the limited therapeutic efficacy of FLT3 inhibitors in FLT3-ITD-positive AML cells and propose feasible solutions. We screened datasets associated with Gilteritinib and Quizartinib in the Gene Expression Omnibus (GEO) database for enrichment analysis and validated potential key pathways that may limit their therapeutic efficacy through qPCR and Western blot. By assessing proliferation, apoptosis, and cell cycle in MV4-11 and MOLM-13 cells, we verified the combined effects of the ERK1/2 inhibitor Ulixertinib with Gilteritinib or Quizartinib, and further explored the underlying mechanisms via transcriptome sequencing. Gilteritinib and Quizartinib both significantly activated the RAS/MAPK pathway in FLT3-ITD-positive AML cells. While the ERK1/2 inhibitor Ulixertinib alone did not inhibit FLT3-ITD-positive AML cells viability, its combination with Gilteritinib or Quizartinib exhibited potent synergistic effects. Transcriptome sequencing revealed that these synergistic effects may stem from the regulation of gene expression such as PKD1, NR2E3, KDF1, and PRSS8 as well as modulation of ion channel activity. This in vitro study identifies aberrant activation of the RAS/MAPK pathway as a critical factor limiting the efficacy of FLT3 inhibitors in FLT3-ITD-positive AML and demonstrates the potent synergistic effects of Ulixertinib combined with FLT3 inhibitors in FLT3-ITD-positive AML cells, providing a novel therapeutic strategy for AML.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"479"},"PeriodicalIF":3.5,"publicationDate":"2025-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145086430","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}