Medical Oncology最新文献

筛选
英文 中文
Effects of APT20TTMG, a modulator of the U1 snRNP complex, in glioblastoma models. U1 snRNP复合体调节剂APT20TTMG在胶质母细胞瘤模型中的作用
IF 3.5 4区 医学
Medical Oncology Pub Date : 2025-10-02 DOI: 10.1007/s12032-025-03057-w
Caio Bruno Quinta de Souza Leal, Camila Guimarães Moreira Zimmer, Vanessa de Vasconcelos Castilho Sinatti, Ericks Sousa Soares, Michael S Rafii, Rafael Mantovani Bottos
{"title":"Effects of APT20TTMG, a modulator of the U1 snRNP complex, in glioblastoma models.","authors":"Caio Bruno Quinta de Souza Leal, Camila Guimarães Moreira Zimmer, Vanessa de Vasconcelos Castilho Sinatti, Ericks Sousa Soares, Michael S Rafii, Rafael Mantovani Bottos","doi":"10.1007/s12032-025-03057-w","DOIUrl":"https://doi.org/10.1007/s12032-025-03057-w","url":null,"abstract":"<p><p>The U1 small nuclear ribonucleoprotein (snRNP) complex is essential for pre-mRNA splicing and inhibition of premature polyadenylation. Its dysfunction has been implicated in various cancers, including glioblastoma, driving oncogenic splicing and tumor progression. This study explored the potential of APT20TTMG, a synthetic cDNA that modulates U1 snRNP misassembly, in glioblastoma. The internalization of APT20TTMG was assessed in U-87 MG cells, as well as its effects on cell viability, proliferation, and apoptosis. Athymic mice were used to evaluate the effects of intravenous APT20TTMG administration on tumor-related parameters. APT20TTMG exhibited over 50% internalization, exerting cytotoxic, cytostatic, and pro-apoptotic effects in vitro. A 22-day treatment with APT20TTMG reduced tumor volume, slowed tumor growth, and showed a trend toward increased body weight. Treatment also decreased oncogenic pathways and tended to enhance histopathological outcomes. A pilot study combining APT20TTMG with temozolomide further improved antitumor efficacy. Our results demonstrate that APT20TTMG has strong potential in correcting U1 snRNP complex dysfunction, supporting its further investigation as a strategy to modulate splicing in glioblastoma.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"507"},"PeriodicalIF":3.5,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145206806","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}
引用次数: 0
Exploring the role of Cathepsin S in mitochondrial energy metabolism: implications for cancer progression and therapeutic targeting. 探索组织蛋白酶S在线粒体能量代谢中的作用:对癌症进展和治疗靶向的影响。
IF 3.5 4区 医学
Medical Oncology Pub Date : 2025-10-01 DOI: 10.1007/s12032-025-03065-w
Rudra Prasad Adhikari, Niladry Sekhar Ghosh
{"title":"Exploring the role of Cathepsin S in mitochondrial energy metabolism: implications for cancer progression and therapeutic targeting.","authors":"Rudra Prasad Adhikari, Niladry Sekhar Ghosh","doi":"10.1007/s12032-025-03065-w","DOIUrl":"https://doi.org/10.1007/s12032-025-03065-w","url":null,"abstract":"<p><p>Cathepsin S (CTSS) is a lysosomal cysteine protease traditionally recognized for its roles in protein degradation and immune responses, but emerging evidence highlights its critical involvement in cancer progression through the regulation of mitochondrial energy metabolism, tumor microenvironment modulation, and apoptosis. CTSS regulates mitochondrial calcium uptake by controlling the mitochondrial calcium uniporter (MCU), thus maintaining mitochondrial membrane potential and oxidative phosphorylation (OXPHOS). Inhibition of CTSS leads to mitochondrial calcium overload, increased reactive oxygen species (ROS) generation, impaired autophagy, and apoptosis, as demonstrated particularly in glioblastoma models. Additionally, CTSS promotes cancer progression by degrading extracellular matrix components, stimulating angiogenesis, and facilitating invasion and metastasis. Selective CTSS inhibitors enhance chemotherapy sensitivity and reduce tumor growth in various preclinical cancer models, including both glycolytic and OXPHOS-dependent tumors. However, most data originate from preclinical studies, limiting immediate clinical applicability. Moreover, CTSS inhibition may elevate ROS levels, posing potential harm to normal cells, and the complex tumor microenvironment presents challenges for targeted therapies. Overall, CTSS is a pivotal regulator that integrates mitochondrial function with tumor microenvironment dynamics, making it a promising therapeutic target. It represents a compelling target for future precision oncology strategies, offering dual benefits of direct tumor suppression and improved sensitivity to existing therapies. Nevertheless, further mechanistic studies and clinical validation are required to fully exploit CTSS's potential in cancer treatment, including deeper investigation into the molecular events linking CTSS inhibition to changes in autophagy, mitochondrial biogenesis, and metabolic reprogramming across diverse cancer subtypes.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"505"},"PeriodicalIF":3.5,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145199789","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}
引用次数: 0
Triple-negative breast cancer: challenges, advances, and promising therapeutic interventions. 三阴性乳腺癌:挑战、进展和有希望的治疗干预措施。
IF 3.5 4区 医学
Medical Oncology Pub Date : 2025-10-01 DOI: 10.1007/s12032-025-03066-9
Deepanshi Sood, Charanjit Kaur, Naresh Kumar, Rajesh Kumar, Gurvinder Singh
{"title":"Triple-negative breast cancer: challenges, advances, and promising therapeutic interventions.","authors":"Deepanshi Sood, Charanjit Kaur, Naresh Kumar, Rajesh Kumar, Gurvinder Singh","doi":"10.1007/s12032-025-03066-9","DOIUrl":"https://doi.org/10.1007/s12032-025-03066-9","url":null,"abstract":"<p><p>Triple-Negative breast cancer (TNBC) is the most aggressive subtype of breast cancer (BC), characterized by absence of estrogen, progesterone, and human epidermal growth factor receptor 2 (HER2). Approximately, 10-15% of BC patients are diagnosed with triple-negative breast cancer. In this review, we have summarized the approaches used for the treatment of triple-negative breast cancer. Due to the complex genetic makeup, Triple-Negative breast cancer has high potential for metastasis and is inherently difficult to treat and poses a grave threat. This review analyzes and provides insight into advancements in treatment methods and diagnostic techniques for TNBC. Schiff base compounds, capable of inducing apoptosis in malignant cells, are effective dual inhibitors of histone deacetylase type II and topoisomerase I. Detection strategies like advanced imaging techniques like core biopsy and automated breast ultrasound are important for enhancing TNBC diagnosis and treatment. The progress in developing combination therapies and improving drug delivery mechanisms has to be maintained in order to tackle the challenges in treatment. This work emphasizes the need for teamwork in the development of therapeutic approaches, which may offer new possibilities in the treatment of TNBC.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"506"},"PeriodicalIF":3.5,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145199801","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}
引用次数: 0
Exosomal transfer of miR-223-3p from carcinoma-associated fibroblasts promotes the malignant properties and chemoresistance of colon cancer cells by targeting NF2/Hippo signaling. 来自癌相关成纤维细胞的miR-223-3p外泌体转移通过靶向NF2/Hippo信号通路促进结肠癌细胞的恶性特性和化疗耐药。
IF 3.5 4区 医学
Medical Oncology Pub Date : 2025-09-30 DOI: 10.1007/s12032-025-03063-y
Jiaqi Zhao, Jiarui Zhang, Jing Liu, Likun Zan
{"title":"Exosomal transfer of miR-223-3p from carcinoma-associated fibroblasts promotes the malignant properties and chemoresistance of colon cancer cells by targeting NF2/Hippo signaling.","authors":"Jiaqi Zhao, Jiarui Zhang, Jing Liu, Likun Zan","doi":"10.1007/s12032-025-03063-y","DOIUrl":"https://doi.org/10.1007/s12032-025-03063-y","url":null,"abstract":"<p><p>Cancer-associated fibroblasts (CAFs) play a key role in malignant progression and chemoresistance of cancer. Accumulating studies indicate that exosomal transfer of microRNAs from CAFs to cancer cells is responsible for the effects of CAFs in cancer (Wang et al. in Cancers, 2021, https://doi.org/10.3390/cancers13133160 ). In the present study, we explored the impact of CAFs-derived exosomes on tumorigenesis and chemoresistance of colon cancer, and potential microRNAs involved in this process. CAFs were isolated form colon cancer samples. CAFs-derived exosomes were separated by ultracentrifugation. Differentially expressed microRNAs were identified by microRNA expression array. The function of CAFs-derived exsomes and exosomal microRNAs were evaluated by cell viability assay, soft agar assay, transwell invasion assay, sphere formation assay, qRT-PCR, tumor xenograft model, flow cytometry, western blot, luciferase reporter assay, biotin microRNA pull-down assay. In our study, CAFs-derived exosomes promoted proliferation, anchorage-independent growth, invasion, stemness, tumor xenograft growth and 5-FU resistance of colon cancer cells. MiR-223-3p was significantly upregulated in CAFs-derived exosomes and serum or tissue samples of colon cancer patients. Exosomal transfer of miR-223-3p facilitated malignant properties and 5-FU resistance of colon cancer cells. Moreover, NF2 was identified as a downstream target for miR-223-3p. Restored NF2 expression partially abrogated the effects caused by exosomal transfer of miR-223-3p in colon cancer cells. In addition, exosomal miR-223-3 regulated Hippo pathway in colon cancer cells by targeting NF2. Our results indicated that exosomal transfer of miR-223-3p from CAFs to colon cancer cells promoted malignant properties and chemoresistance through NF2/Hippo pathway.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"503"},"PeriodicalIF":3.5,"publicationDate":"2025-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145199773","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}
引用次数: 0
Long noncoding RNA RHPN1-AS1 promotes hepatocellular carcinoma progression under hypoxia through interaction with RPS15A protein. 长链非编码RNA rpsn1 - as1通过与RPS15A蛋白相互作用促进缺氧条件下肝细胞癌的进展。
IF 3.5 4区 医学
Medical Oncology Pub Date : 2025-09-30 DOI: 10.1007/s12032-025-03049-w
Qin Peng, Yu-Ting Cai, Qi Ding, Xiang-Yun Qian, Cong Xu, Hang-Cheng Zhou, Hao Chen, Heng Li, Wei Wang
{"title":"Long noncoding RNA RHPN1-AS1 promotes hepatocellular carcinoma progression under hypoxia through interaction with RPS15A protein.","authors":"Qin Peng, Yu-Ting Cai, Qi Ding, Xiang-Yun Qian, Cong Xu, Hang-Cheng Zhou, Hao Chen, Heng Li, Wei Wang","doi":"10.1007/s12032-025-03049-w","DOIUrl":"https://doi.org/10.1007/s12032-025-03049-w","url":null,"abstract":"<p><p>Hypoxic microenvironment is a hallmark feature of hepatocellular carcinoma (HCC) and contributes to cancer progression. RHPN1-AS1, a long noncoding RNA (lncRNA), plays an important role in multiple cancers. However, its expression and oncogenic function under hypoxic conditions have not yet been determined. In this study, we investigated the expression changes of RHPN1-AS1 in HCC cells upon hypoxia. The effects of RHPN1-AS1 knockdown and overexpression on hypoxic HCC cells were explored. The protein partner involved in RHPN1-AS1 action in hypoxic HCC cells was characterized. We found that exposure to hypoxia led to an increase in the RHPN1-AS1 level in HCC cells, which was blocked by depletion of HIF-1α. Chromatin immunoprecipitation assay revealed the enrichment of HIF-1α at the promoter of RHPN1-AS1 in hypoxic HCC cells. Knockdown of RHPN1-AS1 suppressed HCC cell proliferation, colony formation, and invasion under hypoxia, whereas overexpression of RHPN1-AS1 promoted the proliferation and invasion of hypoxic HCC cells. Mechanistically, RHPN1-AS1 interacted with and stabilized RPS15A protein in hypoxic HCC cells. Elevated expression of RPS15A protein enhanced the proliferation and invasion of hypoxic HCC cells through activation of β-catenin signaling. Silencing of RPS15A attenuated RHPN1-AS1-induced aggressiveness and β-catenin activation in hypoxic HCC cells. In vivo tumorigenic studies confirmed that RPS15A depletion significantly reduced the growth of RHPN1-AS1-overexpressing HCC xenograft tumors. RHPN1-AS1 serves as a hypoxia-responsive lncRNA and interacts with the RPS15A protein partner to activate the β-catenin pathway, consequently enhancing HCC progression under hypoxia.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"502"},"PeriodicalIF":3.5,"publicationDate":"2025-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145199830","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}
引用次数: 0
LncRNA LOXL1-AS1 promotes ovarian cancer progression by enhanced BRIP1 mRNA stability. LncRNA LOXL1-AS1通过增强BRIP1 mRNA稳定性促进卵巢癌进展。
IF 3.5 4区 医学
Medical Oncology Pub Date : 2025-09-30 DOI: 10.1007/s12032-025-03055-y
Su Wan, Chang Su, Jin Ding, Ji Liu, Lingli He, Lifen Liu, Qi Peng, Guantai Ni, Weipei Zhu
{"title":"LncRNA LOXL1-AS1 promotes ovarian cancer progression by enhanced BRIP1 mRNA stability.","authors":"Su Wan, Chang Su, Jin Ding, Ji Liu, Lingli He, Lifen Liu, Qi Peng, Guantai Ni, Weipei Zhu","doi":"10.1007/s12032-025-03055-y","DOIUrl":"https://doi.org/10.1007/s12032-025-03055-y","url":null,"abstract":"<p><p>Long non-coding RNAs (lncRNAs) have crucial effects on the development of malignant tumors. This work focused on determining how LOXL1-AS1 contributed to epithelial ovarian cancer development. As indicated by quantitative RT-polymerase chain reaction (qRT-PCR), LOXL1-AS1 showed significant overexpression within ovarian epithelial cancer tissues and ovarian cancer cells compared with non-cancer samples and regular human epithelial cell lines. According to CCK-8, flow cytometry, plate cloning, cell scratch test, a series of cell function tests in vitro, a nude mouse transplanted tumor model, and Western blot assays, LOXL1-AS1 siRNA transfection suppressed the growth, invasion, and epithelial-to-mesenchymal transformation characteristics of SKOV3 and A2780 cells in vitro and vivo. As discovered, LOXL1-AS1 targets BRCA1-interacting protein C-terminal helicase 1 (BRIP1) mRNA, resulting in a malignant phenotype of ovarian cancer. Overexpression of BRIP1 reversed the inhibition of cell progression induced by LOXL1-AS1 siRNA. In addition, based on RNA stability experiments, LOXL1-AS1 enhanced ovarian cancer cell growth and metastasis by stabilizing BRIP1 mRNA. Our findings reveal a novel mechanism of how LOXL1-AS1 enhances epithelial ovarian cancer progression by specifically regulating BRIP1 mRNA stability. This provides the potential therapeutic application of LOXL1-AS1 targeting BRIP1 for treating ovarian cancer.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"504"},"PeriodicalIF":3.5,"publicationDate":"2025-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145199862","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}
引用次数: 0
Hypoxia-drives reversible cell cycle arrest in lung cancer cells via modulation of cellular redox and gene expression. 缺氧通过调节细胞氧化还原和基因表达驱动肺癌细胞的可逆细胞周期阻滞。
IF 3.5 4区 医学
Medical Oncology Pub Date : 2025-09-29 DOI: 10.1007/s12032-025-03058-9
Dharmendra Kumar Maurya, Varshita Mehta, Babita Singh
{"title":"Hypoxia-drives reversible cell cycle arrest in lung cancer cells via modulation of cellular redox and gene expression.","authors":"Dharmendra Kumar Maurya, Varshita Mehta, Babita Singh","doi":"10.1007/s12032-025-03058-9","DOIUrl":"10.1007/s12032-025-03058-9","url":null,"abstract":"<p><p>Hypoxia is a well-recognized clinical feature of solid tumors, including lung cancer, and is associated with poor prognosis due to its role in promoting resistance to chemotherapy and radiotherapy. To investigate the cellular consequences of hypoxia, we cultured A549 lung adenocarcinoma cells under 1% O<sub>2</sub> and examined their growth, cell cycle distribution, and redox status. Hypoxia significantly reduces cell proliferation and induced G1 phase cell cycle arrest, suggesting a cytostatic effect. Biochemical analysis showed a 2.64- and 2.04-fold increase in total and mitochondrial reactive oxygen species (ROS) levels, respectively, along with an elevated total thiol levels under hypoxic conditions compared to normoxia. To assess the reversibility of the hypoxic response, we performed a reciprocal oxygen exposure experiment where cells initially grown under hypoxia were re-exposed to normoxia, and vice versa. To explore the underlying molecular mechanism, we analyzed transcriptomic datasets (GEO accession: GSE48134 and GSE42416) which revealed that hypoxia downregulated key genes involved in energy metabolism (e.g., PDK4, G6PD), cell cycle progression (e.g., CCND1, CDK2), and redox regulation (e.g., GCLM, TXNRD1, NQO1, GCLC). Further, few of redox-related genes were validated by RT-PCR in A549 cells cultured under hypoxia and normoxia for 24 h. Importantly, cyclic hypoxia (intermittent hypoxia-reoxygenation) conditions showed partial restoration of some of these transcripts, supporting the transient nature of hypoxic stress, consistent with our in vitro observations. Furthermore, transcriptome profiles from adenocarcinoma patients (GEO accession: GSE30979) also match our cell line observations. Thus, our results clearly show that hypoxia causes a temporary cell cycle arrest in lung cancer cells, which is reversible when oxygen is restored.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"501"},"PeriodicalIF":3.5,"publicationDate":"2025-09-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12479581/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145192067","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}
引用次数: 0
Engineering adoptive cell therapy for solid tumors. 实体瘤的工程过继细胞治疗。
IF 3.5 4区 医学
Medical Oncology Pub Date : 2025-09-28 DOI: 10.1007/s12032-025-03067-8
Maryam Sanjary, Ameneh Shokati, Mahshid Akhavan Rahnama, Sanaz Khaseb, Mohammad Ahmadvand
{"title":"Engineering adoptive cell therapy for solid tumors.","authors":"Maryam Sanjary, Ameneh Shokati, Mahshid Akhavan Rahnama, Sanaz Khaseb, Mohammad Ahmadvand","doi":"10.1007/s12032-025-03067-8","DOIUrl":"https://doi.org/10.1007/s12032-025-03067-8","url":null,"abstract":"<p><p>Adaptive cell therapy (ACT) has emerged as a promising immunotherapeutic approach for cancer treatment by using engineered immune cells to recognize and destroy malignant cells. While ACT has shown remarkable success in hematologic malignancies, its application in solid tumors remains limited due to unique challenges such as limited immune cell infiltration, antigen heterogeneity, and the immunosuppressive tumor microenvironment. This review provides an overview of current strategies to enhance the efficacy of ACT in solid tumors, focusing on engineered T cells, including CAR-T, TCR-T, and tumor-infiltrating lymphocytes (TILs). We discuss recent progress in cancer immunotherapy, with a focus on tumor targeting, resistance to immunosuppressive signals, as well as strategies to overcome antigen escape. Moreover, we highlight the role of gene-editing tools such as CRISPR/Cas9 in designing next-generation immune cells with enhanced functionality and safety. By integrating novel engineering techniques and systems biology approaches, ACT holds the potential to become a key component of personalized cancer therapy for solid tumors.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"500"},"PeriodicalIF":3.5,"publicationDate":"2025-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145182102","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}
引用次数: 0
Therapeutic implications of vitamin D in leukemia: mechanistic and clinical perspectives. 维生素D在白血病中的治疗意义:机制和临床观点。
IF 3.5 4区 医学
Medical Oncology Pub Date : 2025-09-27 DOI: 10.1007/s12032-025-03052-1
Elham Moasser, Yasin Parvizi, Alireza Samavati, Parmis Taghizadeh, Alireza Moasser, Tahereh Zarei, Mani Ramzi
{"title":"Therapeutic implications of vitamin D in leukemia: mechanistic and clinical perspectives.","authors":"Elham Moasser, Yasin Parvizi, Alireza Samavati, Parmis Taghizadeh, Alireza Moasser, Tahereh Zarei, Mani Ramzi","doi":"10.1007/s12032-025-03052-1","DOIUrl":"https://doi.org/10.1007/s12032-025-03052-1","url":null,"abstract":"<p><p>Recent research has highlighted vitamin D's potential role in the development and treatment of hematological cancers, expanding its known importance beyond calcium regulation and bone health. Patients with blood disorders often exhibit lower serum concentrations of 25(OH)D3, the precursor to calcitriol (1,25(OH)<sub>2</sub>D3, active form of vitamin D), compared to healthy individuals. This deficiency frequently correlates with worse disease outcomes. Additionally, cells affected by these disorders typically show elevated expression of the vitamin D receptor (VDR), which is crucial for many of the observed anti-cancer effects in various experimental models. Supplementation with vitamin D in abnormal blood cells has been shown to encourage cell death, stimulate differentiation, curb proliferation, enhance tumor cell sensitivity to other cancer therapies, and decrease the production of inflammatory cytokines. While the doses required to achieve these effects may lead to hypercalcemia in humans, researchers have developed analogs and combination treatments to mitigate this side effect. These findings underscore the importance of continued research into the role of vitamin D in hematological cancers and its potential as a therapeutic agent or adjuvant in treatment strategies. This review evaluates the role of vitamin D in leukemia by synthesizing mechanistic, clinical, and epidemiological evidence, with the aim of clarifying its prognostic value and therapeutic potential in precision medicine.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"499"},"PeriodicalIF":3.5,"publicationDate":"2025-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145176380","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}
引用次数: 0
Deciphering the anti-cancer and anti-inflammatory activity in natural bioactive compounds of Typhonium flagelliforme: in silico approaches with special target to NEK7. 解读鞭毛霉天然生物活性化合物的抗癌和抗炎活性:以NEK7为特殊靶点的计算机方法。
IF 3.5 4区 医学
Medical Oncology Pub Date : 2025-09-26 DOI: 10.1007/s12032-025-03035-2
Shahanavaj Khan, Salah-Ud-Din Khan, Saeed Vohra, Shahzad Rasheed, Meshari A Alsuwat, Ali Hazazi, Farah Anjum
{"title":"Deciphering the anti-cancer and anti-inflammatory activity in natural bioactive compounds of Typhonium flagelliforme: in silico approaches with special target to NEK7.","authors":"Shahanavaj Khan, Salah-Ud-Din Khan, Saeed Vohra, Shahzad Rasheed, Meshari A Alsuwat, Ali Hazazi, Farah Anjum","doi":"10.1007/s12032-025-03035-2","DOIUrl":"https://doi.org/10.1007/s12032-025-03035-2","url":null,"abstract":"<p><p>Cancer is a great challenge and act as a crucial cause of death globally. Typhonium flagelliforme (T. flagelliforme), a medicinal plant, has been used to treat diverse diseases, including cancer. Consequently, natural compounds from this plant are being investigated for the development of potent and selective anti-cancer agents. A promising target, NEK7 interacts with NLRP3 to assemble the NLRP3 inflammasome critical complex in innate immune responses, and pyroptotic cell death. Altered NEK7 expression is connected to the progression of several cancers, making it a key target for therapy. However, the lack of effective NEK7 inhibitors highlights the urgent need to develop novel therapeutic strategies. The work aimed to identify potential anti-cancer compounds from T. flagelliforme that can act as NEK7 inhibitors for the management of lung cancer. We employed a range of computational techniques such as molecular docking, molecular dynamics (MD) simulations, and binding free energy (MM/PBSA) calculations, to evaluate the plant's bioactive compounds. Molecular docking revealed that several compounds, specifically Beta-Sitosterol, Cycloartane-3.beta.,25-diol, and Ergost-7-en-3-ol, showed higher binding scores -9.5 kcal/mol, -9.2 kcal/mol, and -9.4 kcal/mol, respectively, than the known reference inhibitor, F9N, which has binding scores of -9.1 kcal/mol. Further analysis through molecular dynamics simulations confirmed that Beta-Sitosterol, Cycloartane-3.beta.,25-diol, and Ergost-7-en-3-ol exhibit a strong and stable binding potential with NEK7. Moreover, binding free energy calculations showed that Cycloartane-3.beta.,25-diol has the highest binding free energy of -203.460 kJ/mol than the positive control (F9N), which has the binding free energy of -170.420 kJ/mol, strongly suggesting its effectiveness in modulating NEK7 signaling. Overall, this research shows that the compounds Cycloartane-3.beta., 25-diol and Beta-Sitosterol have great potential to inhibit NEK7, surpassing the known F9N compound. The study emphasizes the therapeutic potential of active compounds from T. flagelliforme for developing new cancer treatments.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 11","pages":"495"},"PeriodicalIF":3.5,"publicationDate":"2025-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145149834","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}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信