首页 > 最新文献

Medical Oncology最新文献

英文 中文
KLHL4 upregulates EGFR signaling to promote the malignant progression of oral squamous cell carcinoma. KLHL4上调EGFR信号,促进口腔鳞状细胞癌的恶性进展。
IF 3.5 4区 医学 Q2 ONCOLOGY Pub Date : 2025-12-26 DOI: 10.1007/s12032-025-03167-5
Yingxin Zhang, Yuan Ren, Yimei Wang, Ming Liu, Fangbu Cheng, Haiyun Li, Silu Sun, Xikun Zhou, Jiantang Yang, Jing Li

Oral squamous cell carcinoma (OSCC) is the most prevalent form of head and neck squamous cell carcinoma (HNSCC), characterized by high incidence rates, frequent recurrence and metastasis, and low survival rates. KLHL4, a member of the Kelch-like family proteins, plays a significant role in cancer. However, the role of KLHL4 in OSCC remains largely unexplored. In this study, we first identified the aberrant overexpression of KLHL4 in OSCC tissues through Western blotting and immunohistochemistry. Subsequent experiments, including qPCR, colony formation assays, scratch assays, and transwell invasion and migration assays, demonstrated that knockdown of KLHL4 inhibits OSCC growth, migration and invasion in vitro. Furthermore, through the establishment of subcutaneous xenograft models and lung metastasis models in nude mice, we revealed that KLHL4 knockdown suppresses tumor growth and metastasis in vivo. We also found that KLHL4 knockout inhibited 4NQO-induced OSCC progression. Mechanistically, co-immunoprecipitation confirmed the physical interaction between KLHL4 and EGFR, while rescue experiments using EGF and Afatinib demonstrated the functional dependency of KLHL4 on EGFR pathway activation. Clinical analysis of a 112-case OSCC tissue microarray revealed that high KLHL4 expression correlated significantly with lymph node metastasis and predicted poor patient overall survival, which was independently validated in a public HNSCC cohort. Overall, our research highlights the critical role of KLHL4 in the malignant progression of OSCC through upregulating the EGFR signaling pathway, indicating its potential as a therapeutic target for OSCC treatment.

口腔鳞状细胞癌(Oral squamous cell carcinoma, OSCC)是头颈部鳞状细胞癌(HNSCC)中最常见的一种,具有发病率高、复发转移频繁、生存率低的特点。KLHL4是kelch样家族蛋白的一员,在癌症中起着重要作用。然而,KLHL4在OSCC中的作用在很大程度上仍未被探索。在本研究中,我们首先通过Western blotting和免疫组织化学方法发现了KLHL4在OSCC组织中的异常过表达。随后的实验,包括qPCR、菌落形成试验、划痕试验和跨井侵袭和迁移试验,表明敲低KLHL4可抑制OSCC的生长、迁移和体外侵袭。此外,通过建立裸鼠皮下异种移植模型和肺转移模型,我们发现KLHL4敲低可抑制体内肿瘤的生长和转移。我们还发现,敲除KLHL4抑制4nqo诱导的OSCC进展。机制上,共免疫沉淀证实了KLHL4与EGFR之间的物理相互作用,而使用EGF和阿法替尼的救援实验证实了KLHL4对EGFR通路激活的功能依赖性。对112例OSCC组织芯片的临床分析显示,高KLHL4表达与淋巴结转移显著相关,并预测患者总生存期较差,这在一项公开的HNSCC队列中得到了独立验证。总之,我们的研究强调了KLHL4通过上调EGFR信号通路在OSCC恶性进展中的关键作用,表明其作为OSCC治疗的治疗靶点的潜力。
{"title":"KLHL4 upregulates EGFR signaling to promote the malignant progression of oral squamous cell carcinoma.","authors":"Yingxin Zhang, Yuan Ren, Yimei Wang, Ming Liu, Fangbu Cheng, Haiyun Li, Silu Sun, Xikun Zhou, Jiantang Yang, Jing Li","doi":"10.1007/s12032-025-03167-5","DOIUrl":"https://doi.org/10.1007/s12032-025-03167-5","url":null,"abstract":"<p><p>Oral squamous cell carcinoma (OSCC) is the most prevalent form of head and neck squamous cell carcinoma (HNSCC), characterized by high incidence rates, frequent recurrence and metastasis, and low survival rates. KLHL4, a member of the Kelch-like family proteins, plays a significant role in cancer. However, the role of KLHL4 in OSCC remains largely unexplored. In this study, we first identified the aberrant overexpression of KLHL4 in OSCC tissues through Western blotting and immunohistochemistry. Subsequent experiments, including qPCR, colony formation assays, scratch assays, and transwell invasion and migration assays, demonstrated that knockdown of KLHL4 inhibits OSCC growth, migration and invasion in vitro. Furthermore, through the establishment of subcutaneous xenograft models and lung metastasis models in nude mice, we revealed that KLHL4 knockdown suppresses tumor growth and metastasis in vivo. We also found that KLHL4 knockout inhibited 4NQO-induced OSCC progression. Mechanistically, co-immunoprecipitation confirmed the physical interaction between KLHL4 and EGFR, while rescue experiments using EGF and Afatinib demonstrated the functional dependency of KLHL4 on EGFR pathway activation. Clinical analysis of a 112-case OSCC tissue microarray revealed that high KLHL4 expression correlated significantly with lymph node metastasis and predicted poor patient overall survival, which was independently validated in a public HNSCC cohort. Overall, our research highlights the critical role of KLHL4 in the malignant progression of OSCC through upregulating the EGFR signaling pathway, indicating its potential as a therapeutic target for OSCC treatment.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"43 2","pages":"70"},"PeriodicalIF":3.5,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145834302","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
Intelligent design of tumor microenvironment-responsive Adeno-associated virus vectors: overcoming delivery barriers and enabling precision therapy. 肿瘤微环境响应性腺相关病毒载体的智能设计:克服传递障碍,实现精准治疗。
IF 3.5 4区 医学 Q2 ONCOLOGY Pub Date : 2025-12-26 DOI: 10.1007/s12032-025-03158-6
Ziyi Wu, Hongmei Liu, Decheng Wu

Adeno-associated virus (AAV) has emerged as a pivotal vector for cancer gene therapy due to its low immunogenicity, non-pathogenicity, and sustained transgene expression capacity. However, the heterogeneity and complexity of the tumor microenvironment (TME) significantly constrain the delivery efficiency and targeting precision of AAV in solid tumors. Dense extracellular matrix, acidic and hypoxic conditions, and immunosuppressive signaling networks collectively impede effective AAV transduction while increasing off-target risks. To overcome these barriers, recent advances have introduced interdisciplinary optimization strategies, including dynamic engineering of AAV capsids, TME-responsive gene expression systems, and biomimetic camouflage technologies to enhance immune evasion and tumor targeting. Furthermore, data-driven AAV engineering, integrating machine learning and high-throughput screening, has significantly accelerated the development of next-generation vectors. This review systematically summarizes intelligent design strategies for TME-responsive AAV vectors and their progress in precision oncology, with a focus on overcoming key delivery barriers to achieve highly efficient and low-toxicity cancer therapy.

腺相关病毒(AAV)由于其低免疫原性、无致病性和持续的转基因表达能力而成为癌症基因治疗的关键载体。然而,肿瘤微环境(tumor microenvironment, TME)的异质性和复杂性极大地限制了AAV在实体肿瘤中的递送效率和靶向精度。致密的细胞外基质、酸性和缺氧条件以及免疫抑制信号网络共同阻碍了AAV的有效转导,同时增加了脱靶风险。为了克服这些障碍,最近的进展引入了跨学科的优化策略,包括AAV衣壳的动态工程、tme响应基因表达系统和仿生伪装技术,以增强免疫逃避和肿瘤靶向。此外,数据驱动的AAV工程,集成了机器学习和高通量筛选,大大加速了下一代载体的发展。本文系统总结了tme响应型AAV载体的智能设计策略及其在精准肿瘤学领域的进展,重点介绍了如何克服关键的递送障碍,实现高效低毒的癌症治疗。
{"title":"Intelligent design of tumor microenvironment-responsive Adeno-associated virus vectors: overcoming delivery barriers and enabling precision therapy.","authors":"Ziyi Wu, Hongmei Liu, Decheng Wu","doi":"10.1007/s12032-025-03158-6","DOIUrl":"https://doi.org/10.1007/s12032-025-03158-6","url":null,"abstract":"<p><p>Adeno-associated virus (AAV) has emerged as a pivotal vector for cancer gene therapy due to its low immunogenicity, non-pathogenicity, and sustained transgene expression capacity. However, the heterogeneity and complexity of the tumor microenvironment (TME) significantly constrain the delivery efficiency and targeting precision of AAV in solid tumors. Dense extracellular matrix, acidic and hypoxic conditions, and immunosuppressive signaling networks collectively impede effective AAV transduction while increasing off-target risks. To overcome these barriers, recent advances have introduced interdisciplinary optimization strategies, including dynamic engineering of AAV capsids, TME-responsive gene expression systems, and biomimetic camouflage technologies to enhance immune evasion and tumor targeting. Furthermore, data-driven AAV engineering, integrating machine learning and high-throughput screening, has significantly accelerated the development of next-generation vectors. This review systematically summarizes intelligent design strategies for TME-responsive AAV vectors and their progress in precision oncology, with a focus on overcoming key delivery barriers to achieve highly efficient and low-toxicity cancer therapy.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"43 2","pages":"66"},"PeriodicalIF":3.5,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145834329","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
Ultrasound-Stimulated microbubble cavitation improved tumor perfusion and promoted tumor vascular normalization in a rabbit VX2 tumor model. 超声刺激的微泡空化改善兔VX2肿瘤模型的肿瘤灌注,促进肿瘤血管正常化。
IF 3.5 4区 医学 Q2 ONCOLOGY Pub Date : 2025-12-26 DOI: 10.1007/s12032-025-03226-x
Tingting Luo, Luhua Bai, Lei Yao, Yi Zhang, Guoliang Yang, Leidan Huang, Xiaoqin Chen, Zheng Liu

Tumor hypoxia poses a major challenge in tumor therapy. Many strategies have been explored to alleviate tumor hypoxic microenvironment to improve the efficacy of tumor therapy. Ultrasound-stimulated microbubbles cavitation (USMC) was proved to improve tumor perfusion, and thus to alleviate tumor hypoxia. The synergistic role of USMC in tumor therapy has been identified by several preclinical and clinical studies. The effect of USMC on improving tumor perfusion is influenced by many factors, and the stability and reproducibility of this effect in long-term tumor treatment remain to be explored. In this study, we established rabbit VX2 tumor model. Fifteen tumor-bearing rabbits were enrolled to compare the effects of USMC with two different mechanical indexes (MIs). The results of contrast-enhanced ultrasound (CEUS) imaging showed that USMC with MI of 0.25 could improve the tumor perfusion better compared with MI 0.40. Then we conducted repeated USMC treatments on tumor-bearing rabbits once a week for six weeks. The results of CEUS showed that USMC with appropriate parameters could always enhance the tumor perfusion although the tumor had developed. Transmission electron microscopy revealed that tumors received multiple USMC treatments had more integral vascular structures compared with the control. Immunofluorescence staining showed that tumors received multiple USMC treatments had higher overlap coefficient of CD31 and intercellular cell adhesion molecule-1, indicating the normalization of tumor vasculature. In conclusion, USMC with appropriate parameters has stability and reproducibility in improving tumor perfusion. And multiple USMC treatments could potentially promote tumor vascular normalization, which is beneficial for tumor therapy.

肿瘤缺氧是肿瘤治疗的主要挑战。为了改善肿瘤缺氧微环境,提高肿瘤治疗效果,人们探索了多种策略。超声刺激微泡空化(USMC)可改善肿瘤灌注,从而缓解肿瘤缺氧。USMC在肿瘤治疗中的协同作用已经被一些临床前和临床研究确定。USMC改善肿瘤灌注的作用受多种因素影响,其在长期肿瘤治疗中的稳定性和可重复性有待探索。本研究建立兔VX2肿瘤模型。选取荷瘤兔15只,比较两种不同力学指标(MIs)对USMC的影响。超声造影(CEUS)结果显示,与MI为0.40的USMC相比,MI为0.25的USMC能更好地改善肿瘤灌注。然后对荷瘤家兔进行USMC治疗,每周1次,连续6周。超声造影结果显示,尽管肿瘤已发生,但适当参数的USMC仍能增强肿瘤灌注。透射电镜显示,与对照组相比,接受多次USMC治疗的肿瘤具有更完整的血管结构。免疫荧光染色显示,多次USMC处理的肿瘤CD31和细胞间细胞粘附分子-1的重叠系数较高,表明肿瘤血管系统正常化。综上所述,适当参数的USMC在改善肿瘤灌注方面具有稳定性和重复性。多次USMC治疗可潜在促进肿瘤血管正常化,有利于肿瘤治疗。
{"title":"Ultrasound-Stimulated microbubble cavitation improved tumor perfusion and promoted tumor vascular normalization in a rabbit VX2 tumor model.","authors":"Tingting Luo, Luhua Bai, Lei Yao, Yi Zhang, Guoliang Yang, Leidan Huang, Xiaoqin Chen, Zheng Liu","doi":"10.1007/s12032-025-03226-x","DOIUrl":"https://doi.org/10.1007/s12032-025-03226-x","url":null,"abstract":"<p><p>Tumor hypoxia poses a major challenge in tumor therapy. Many strategies have been explored to alleviate tumor hypoxic microenvironment to improve the efficacy of tumor therapy. Ultrasound-stimulated microbubbles cavitation (USMC) was proved to improve tumor perfusion, and thus to alleviate tumor hypoxia. The synergistic role of USMC in tumor therapy has been identified by several preclinical and clinical studies. The effect of USMC on improving tumor perfusion is influenced by many factors, and the stability and reproducibility of this effect in long-term tumor treatment remain to be explored. In this study, we established rabbit VX2 tumor model. Fifteen tumor-bearing rabbits were enrolled to compare the effects of USMC with two different mechanical indexes (MIs). The results of contrast-enhanced ultrasound (CEUS) imaging showed that USMC with MI of 0.25 could improve the tumor perfusion better compared with MI 0.40. Then we conducted repeated USMC treatments on tumor-bearing rabbits once a week for six weeks. The results of CEUS showed that USMC with appropriate parameters could always enhance the tumor perfusion although the tumor had developed. Transmission electron microscopy revealed that tumors received multiple USMC treatments had more integral vascular structures compared with the control. Immunofluorescence staining showed that tumors received multiple USMC treatments had higher overlap coefficient of CD31 and intercellular cell adhesion molecule-1, indicating the normalization of tumor vasculature. In conclusion, USMC with appropriate parameters has stability and reproducibility in improving tumor perfusion. And multiple USMC treatments could potentially promote tumor vascular normalization, which is beneficial for tumor therapy.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"43 2","pages":"89"},"PeriodicalIF":3.5,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145834376","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 potential of natural triterpenoids in liver cancer. 天然三萜对肝癌的治疗潜力。
IF 3.5 4区 医学 Q2 ONCOLOGY Pub Date : 2025-12-26 DOI: 10.1007/s12032-025-03155-9
Chengu Niu, Jing Zhang, Patrick I Okolo Iii

Pathologically, primary liver cancer is a heterogeneous, aggressive malignancy and encompasses hepatocellular carcinoma, intrahepatic cholangiocarcinoma, and other rare tumors. This cancer remains the fifth most common malignancy in men and the eighth in women and the fourth leading cause of cancer-related mortality worldwide. Clinically, most liver cancer occurs typically from a background of cirrhosis and chronic inflammation. Liver cancer remains difficult to treat; small-molecule kinase inhibitors such as sorafenib and lenvatinib have only limited clinical benefit. Therefore, an urgent need exists to develop novel drugs for the treatment of liver cancer. Triterpenoids, consisting of six isoprene units, are structurally diverse secondary metabolites and possess versatile biological activities including immunomodulatory and antitumor activities. In this review, we discuss the therapeutic potential of natural triterpenoids in liver cancer, primarily focusing on their possible mechanisms involving induction of cell cycle arrest, initiation of apoptosis, activation of autophagy, stimulation of antitumor immune response, suppression of angiogenesis and metastasis, and enhancement of radio/chemo-sensitivity. We also present the current challenges that impede their translation to clinical practices. We conclude that natural triterpenoids could serve as potential candidates for developing liver cancer therapies, based on the current preclinical evidence. However, their clinical translation requires further validation. Hopefully, the knowledge gained from this review will outline future research directions and identify current research priorities in this field.

病理上,原发性肝癌是一种异质性、侵袭性的恶性肿瘤,包括肝细胞癌、肝内胆管癌和其他罕见肿瘤。这种癌症仍然是男性第五大最常见恶性肿瘤,女性第八大最常见恶性肿瘤,也是全球癌症相关死亡的第四大原因。临床上,大多数肝癌通常发生在肝硬化和慢性炎症的背景下。肝癌仍然难以治疗;小分子激酶抑制剂如索拉非尼和lenvatinib只有有限的临床益处。因此,迫切需要开发治疗肝癌的新药。三萜由6个异戊二烯单位组成,是结构多样的次生代谢产物,具有多种生物活性,包括免疫调节和抗肿瘤活性。在这篇综述中,我们讨论了天然三萜在肝癌中的治疗潜力,主要关注其可能的机制,包括诱导细胞周期阻滞,启动细胞凋亡,激活自噬,刺激抗肿瘤免疫反应,抑制血管生成和转移,以及增强放射/化学敏感性。我们还提出了当前阻碍其转化为临床实践的挑战。我们得出结论,基于目前的临床前证据,天然三萜可以作为开发肝癌治疗的潜在候选者。然而,它们的临床转化需要进一步的验证。希望从这篇综述中获得的知识将概述未来的研究方向,并确定当前该领域的研究重点。
{"title":"Therapeutic potential of natural triterpenoids in liver cancer.","authors":"Chengu Niu, Jing Zhang, Patrick I Okolo Iii","doi":"10.1007/s12032-025-03155-9","DOIUrl":"https://doi.org/10.1007/s12032-025-03155-9","url":null,"abstract":"<p><p>Pathologically, primary liver cancer is a heterogeneous, aggressive malignancy and encompasses hepatocellular carcinoma, intrahepatic cholangiocarcinoma, and other rare tumors. This cancer remains the fifth most common malignancy in men and the eighth in women and the fourth leading cause of cancer-related mortality worldwide. Clinically, most liver cancer occurs typically from a background of cirrhosis and chronic inflammation. Liver cancer remains difficult to treat; small-molecule kinase inhibitors such as sorafenib and lenvatinib have only limited clinical benefit. Therefore, an urgent need exists to develop novel drugs for the treatment of liver cancer. Triterpenoids, consisting of six isoprene units, are structurally diverse secondary metabolites and possess versatile biological activities including immunomodulatory and antitumor activities. In this review, we discuss the therapeutic potential of natural triterpenoids in liver cancer, primarily focusing on their possible mechanisms involving induction of cell cycle arrest, initiation of apoptosis, activation of autophagy, stimulation of antitumor immune response, suppression of angiogenesis and metastasis, and enhancement of radio/chemo-sensitivity. We also present the current challenges that impede their translation to clinical practices. We conclude that natural triterpenoids could serve as potential candidates for developing liver cancer therapies, based on the current preclinical evidence. However, their clinical translation requires further validation. Hopefully, the knowledge gained from this review will outline future research directions and identify current research priorities in this field.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"43 2","pages":"87"},"PeriodicalIF":3.5,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145834391","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
Anti-tumor effects and mechanism of the histone demethylase inhibitor GSK-J4 in non-small cell lung cancer cells. 组蛋白去甲基化酶抑制剂GSK-J4在非小细胞肺癌细胞中的抗肿瘤作用及机制
IF 3.5 4区 医学 Q2 ONCOLOGY Pub Date : 2025-12-26 DOI: 10.1007/s12032-025-03185-3
Dandan Xu, Menghan Wang, Mingyuan Wu, Yao Yu, Xingxuan Chen, Zhe Wen, Danting Shen, Tao Liu, Hong Chen

Lung cancer is one of the leading causes of death worldwide. Cancer metastases are responsible for 90% of cancer-related deaths. In view of the high incidence and mortality of lung cancer, there is still an urgent clinical need to improve the early diagnosis of lung cancer and explore new therapeutic methods and targets to improve the prognosis of patients. By targeting promoter or enhancer regions of related genes, histone methylation modification dynamically regulates gene expression and activation of signaling pathways, and is involved in mediating malignant biological processes such as proliferation, invasion and metastasis of tumor cells. GSK-J4 is a novel small molecule inhibitor of the JMJD3 and UTX family of selective histone demethylases, which shows good anticancer activity against various types of tumors by inhibiting H3K27me3 demethylation. The aim of this study was to investigate the effects of GSK-J4 on proliferation, migration, invasion and epithelial-mesenchymal transformation (EMT) of TGF-β1-induced non-small cell lung cancer (NSCLC) cell lines A549 and H1299. The effect of GSK-J4 on cell proliferation was detected by CCK8, clonal formation assay, immunofluorescence and flow cytometry. The effects of GSK-J4 on the migration and invasion of A549 and H1299 cells induced by TGFβ1 were examined by wound healing assay, Transwell migration assay, and then, the expression changes of related markers were detected by RT-qPCR and western blot. Finally, GSK-J4 was verified to inhibit tumor growth in vivo by constructing a mouse model of tumor implantation in situ, and observed its effectiveness and safety. GSK-J4 inhibited proliferation and promoted apoptosis of A549 and H1299. GSK-J4 inhibited EMT, invasion and migration of TGFβ1-induced NSCLC. GSK-J4 inhibits EMT, invasion and migration of TGFβ1-induced NSCLC through the classical Wnt/β-catenin signaling pathway. In situ tumor model, GSK-J4 administration alone effectively reduced tumor growth in nude mice, with the tumor size being notably less than in the control group. IHC analysis showed that the expression of Ki-67 in GSK-J4 administration group was lower than that in control group. HE staining showed that GSK-J4 had no significant effect on the histopathology of heart, liver, lung, kidney and other major organs of mice. GSK-J4, an inhibitor of histone demethylase, elevated the level of H3K27me3 by suppressing JMJD3/UTX activity, thus curbing NSCLC cell growth and encouraging cell death. Concurrently, GSK-J4 played a role in preventing TGFβ1-triggered EMT, invasion, and migration. Consequently, targeting histone methylation modification and the small molecule inhibitor GSK-J4 is anticipated to be an effective treatment strategy and a novel method for NSCLC.

肺癌是世界范围内导致死亡的主要原因之一。90%的癌症相关死亡是由癌症转移引起的。鉴于肺癌的高发病率和高死亡率,临床仍迫切需要提高肺癌的早期诊断,探索新的治疗方法和靶点,以改善患者的预后。组蛋白甲基化修饰通过靶向相关基因的启动子或增强子区域,动态调控基因表达和信号通路激活,参与介导肿瘤细胞增殖、侵袭、转移等恶性生物学过程。GSK-J4是JMJD3和UTX家族选择性组蛋白去甲基化酶的新型小分子抑制剂,通过抑制H3K27me3去甲基化,对多种类型肿瘤显示出良好的抗癌活性。本研究旨在探讨GSK-J4对TGF-β1诱导的非小细胞肺癌(NSCLC)细胞株A549、H1299增殖、迁移、侵袭及上皮间充质转化(EMT)的影响。采用CCK8、克隆形成实验、免疫荧光和流式细胞术检测GSK-J4对细胞增殖的影响。采用创面愈合实验、Transwell迁移实验检测GSK-J4对tgf - β1诱导的A549和H1299细胞迁移和侵袭的影响,并采用RT-qPCR和western blot检测相关标志物的表达变化。最后,通过构建小鼠原位肿瘤植入模型,验证GSK-J4在体内具有抑制肿瘤生长的作用,并观察其有效性和安全性。GSK-J4抑制A549和H1299细胞增殖,促进细胞凋亡。GSK-J4抑制tgf β1诱导的非小细胞肺癌的EMT、侵袭和迁移。GSK-J4通过经典的Wnt/β-catenin信号通路抑制tgf - β1诱导的NSCLC的EMT、侵袭和迁移。原位肿瘤模型中,单独给药GSK-J4能有效抑制裸鼠肿瘤生长,肿瘤大小明显小于对照组。免疫组化分析显示,GSK-J4给药组Ki-67的表达低于对照组。HE染色显示GSK-J4对小鼠心、肝、肺、肾等主要器官的组织病理学无明显影响。GSK-J4是一种组蛋白去甲基化酶抑制剂,通过抑制JMJD3/UTX活性提高H3K27me3水平,从而抑制NSCLC细胞生长并促进细胞死亡。同时,GSK-J4在阻止tgf β1触发的EMT、侵袭和迁移中发挥作用。因此,靶向组蛋白甲基化修饰和小分子抑制剂GSK-J4有望成为一种有效的治疗策略和治疗NSCLC的新方法。
{"title":"Anti-tumor effects and mechanism of the histone demethylase inhibitor GSK-J4 in non-small cell lung cancer cells.","authors":"Dandan Xu, Menghan Wang, Mingyuan Wu, Yao Yu, Xingxuan Chen, Zhe Wen, Danting Shen, Tao Liu, Hong Chen","doi":"10.1007/s12032-025-03185-3","DOIUrl":"10.1007/s12032-025-03185-3","url":null,"abstract":"<p><p>Lung cancer is one of the leading causes of death worldwide. Cancer metastases are responsible for 90% of cancer-related deaths. In view of the high incidence and mortality of lung cancer, there is still an urgent clinical need to improve the early diagnosis of lung cancer and explore new therapeutic methods and targets to improve the prognosis of patients. By targeting promoter or enhancer regions of related genes, histone methylation modification dynamically regulates gene expression and activation of signaling pathways, and is involved in mediating malignant biological processes such as proliferation, invasion and metastasis of tumor cells. GSK-J4 is a novel small molecule inhibitor of the JMJD3 and UTX family of selective histone demethylases, which shows good anticancer activity against various types of tumors by inhibiting H3K27me3 demethylation. The aim of this study was to investigate the effects of GSK-J4 on proliferation, migration, invasion and epithelial-mesenchymal transformation (EMT) of TGF-β1-induced non-small cell lung cancer (NSCLC) cell lines A549 and H1299. The effect of GSK-J4 on cell proliferation was detected by CCK8, clonal formation assay, immunofluorescence and flow cytometry. The effects of GSK-J4 on the migration and invasion of A549 and H1299 cells induced by TGFβ1 were examined by wound healing assay, Transwell migration assay, and then, the expression changes of related markers were detected by RT-qPCR and western blot. Finally, GSK-J4 was verified to inhibit tumor growth in vivo by constructing a mouse model of tumor implantation in situ, and observed its effectiveness and safety. GSK-J4 inhibited proliferation and promoted apoptosis of A549 and H1299. GSK-J4 inhibited EMT, invasion and migration of TGFβ1-induced NSCLC. GSK-J4 inhibits EMT, invasion and migration of TGFβ1-induced NSCLC through the classical Wnt/β-catenin signaling pathway. In situ tumor model, GSK-J4 administration alone effectively reduced tumor growth in nude mice, with the tumor size being notably less than in the control group. IHC analysis showed that the expression of Ki-67 in GSK-J4 administration group was lower than that in control group. HE staining showed that GSK-J4 had no significant effect on the histopathology of heart, liver, lung, kidney and other major organs of mice. GSK-J4, an inhibitor of histone demethylase, elevated the level of H3K27me3 by suppressing JMJD3/UTX activity, thus curbing NSCLC cell growth and encouraging cell death. Concurrently, GSK-J4 played a role in preventing TGFβ1-triggered EMT, invasion, and migration. Consequently, targeting histone methylation modification and the small molecule inhibitor GSK-J4 is anticipated to be an effective treatment strategy and a novel method for NSCLC.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"43 2","pages":"86"},"PeriodicalIF":3.5,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12743077/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145833923","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
"Exosomal blueprint of lung-tropic metastasis: molecular signatures, microenvironmental conditioning, and translational implications in cancer". 嗜肺转移的外泌体蓝图:分子特征、微环境调节和癌症的翻译意义。
IF 3.5 4区 医学 Q2 ONCOLOGY Pub Date : 2025-12-26 DOI: 10.1007/s12032-025-03217-y
Noura A A Ebrahim, Thoraya A Farghaly, Soliman M A Soliman

Exosomes are increasingly recognized as central regulators of organ-specific metastasis, and this review concentrates on their contribution to lung-directed dissemination. Yet, a detailed and integrative synthesis of how exosomes contribute to lung-directed metastatic spread-and what these mechanisms mean for clinical translation-remains largely absent from the current literature. Extracellular vesicles (EVs), particularly exosomes measuring 30-150 nm, are nanoscale, lipid bilayer-enclosed structures secreted by nearly all cell types. In cancer, tumor-derived exosomes act as potent mediators of intercellular signaling, enabling metastatic spread by modulating inflammation, angiogenesis, extracellular matrix dynamics, and immune evasion. Their molecular cargo, especially integrin profiles, plays a decisive role in determining metastatic tropism: integrins α6β4 and α6β1 are strongly associated with pulmonary colonization, while αvβ5 directs metastasis toward the liver. In malignancies such as breast, colorectal, melanoma, and pancreatic cancer, exosomal proteins and RNAs remodel the lung microenvironment, enhancing vascular permeability and attracting stromal and immune components that establish a receptive pre-metastatic niche. Clinically, exosomes are emerging as powerful liquid biopsy biomarkers and as promising platforms for targeted drug delivery. Advances in EV bioengineering now permit tailoring of surface molecules and cargo to improve pulmonary selectivity, for instance, supporting selective delivery of therapeutic payloads to pulmonary tumors or altering immune dynamics within the lung microenvironment, while omics-based and imaging technologies support detailed profiling and tracking. Early clinical trials of exosome-derived vaccines and therapeutic carriers have demonstrated feasibility and safety, although no EV-based therapy has yet achieved regulatory approval. This review integrates mechanistic insights, niche biology, and translational advances to highlight the unique role of exosomes in lung-specific metastasis and their potential as diagnostic and therapeutic tools in precision oncology.

外泌体越来越被认为是器官特异性转移的中枢调节因子,本文主要讨论它们在肺定向转移中的作用。然而,关于外泌体如何促进肺转移扩散以及这些机制对临床翻译的意义的详细和综合的综合,在目前的文献中仍然很大程度上缺乏。细胞外囊泡(EVs)是一种纳米级的脂质双层封闭结构,几乎所有细胞类型都有分泌,尤其是直径30-150 nm的外泌体。在癌症中,肿瘤源性外泌体作为细胞间信号传导的有效介质,通过调节炎症、血管生成、细胞外基质动力学和免疫逃避来实现转移扩散。它们的分子载体,尤其是整合素谱,在决定转移倾向中起决定性作用:整合素α6β4和α6β1与肺定植密切相关,而αvβ5则引导转移到肝脏。在乳腺癌、结直肠癌、黑色素瘤和胰腺癌等恶性肿瘤中,外泌体蛋白和rna重塑肺微环境,增强血管通透性,吸引基质和免疫成分,建立接受性转移前生态位。在临床上,外泌体正在成为强有力的液体活检生物标志物和有希望的靶向药物递送平台。EV生物工程的进步现在允许定制表面分子和货物以提高肺选择性,例如,支持选择性地向肺肿瘤提供治疗有效载荷或改变肺微环境中的免疫动力学,而基于组学和成像技术支持详细的分析和跟踪。外泌体衍生疫苗和治疗性载体的早期临床试验已经证明了可行性和安全性,尽管尚未有基于外泌体的疗法获得监管部门的批准。本文综述了外泌体在肺特异性转移中的独特作用,以及它们作为精确肿瘤学诊断和治疗工具的潜力。
{"title":"\"Exosomal blueprint of lung-tropic metastasis: molecular signatures, microenvironmental conditioning, and translational implications in cancer\".","authors":"Noura A A Ebrahim, Thoraya A Farghaly, Soliman M A Soliman","doi":"10.1007/s12032-025-03217-y","DOIUrl":"https://doi.org/10.1007/s12032-025-03217-y","url":null,"abstract":"<p><p>Exosomes are increasingly recognized as central regulators of organ-specific metastasis, and this review concentrates on their contribution to lung-directed dissemination. Yet, a detailed and integrative synthesis of how exosomes contribute to lung-directed metastatic spread-and what these mechanisms mean for clinical translation-remains largely absent from the current literature. Extracellular vesicles (EVs), particularly exosomes measuring 30-150 nm, are nanoscale, lipid bilayer-enclosed structures secreted by nearly all cell types. In cancer, tumor-derived exosomes act as potent mediators of intercellular signaling, enabling metastatic spread by modulating inflammation, angiogenesis, extracellular matrix dynamics, and immune evasion. Their molecular cargo, especially integrin profiles, plays a decisive role in determining metastatic tropism: integrins α6β4 and α6β1 are strongly associated with pulmonary colonization, while αvβ5 directs metastasis toward the liver. In malignancies such as breast, colorectal, melanoma, and pancreatic cancer, exosomal proteins and RNAs remodel the lung microenvironment, enhancing vascular permeability and attracting stromal and immune components that establish a receptive pre-metastatic niche. Clinically, exosomes are emerging as powerful liquid biopsy biomarkers and as promising platforms for targeted drug delivery. Advances in EV bioengineering now permit tailoring of surface molecules and cargo to improve pulmonary selectivity, for instance, supporting selective delivery of therapeutic payloads to pulmonary tumors or altering immune dynamics within the lung microenvironment, while omics-based and imaging technologies support detailed profiling and tracking. Early clinical trials of exosome-derived vaccines and therapeutic carriers have demonstrated feasibility and safety, although no EV-based therapy has yet achieved regulatory approval. This review integrates mechanistic insights, niche biology, and translational advances to highlight the unique role of exosomes in lung-specific metastasis and their potential as diagnostic and therapeutic tools in precision oncology.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"43 2","pages":"93"},"PeriodicalIF":3.5,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145833967","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
Fatty acid metabolic signature reveals FASN as an immunosuppressive factor in DLBCL tumor microenvironment. 脂肪酸代谢特征显示FASN是DLBCL肿瘤微环境中的免疫抑制因子。
IF 3.5 4区 医学 Q2 ONCOLOGY Pub Date : 2025-12-26 DOI: 10.1007/s12032-025-03159-5
Tingting Cheng, Shiyu Wang, Yue Zhang, Fenfang Tong, Wei Qin, Yajing Xu

Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin lymphoma (NHL). Targeting fatty acid metabolism pathway represents a promising therapeutic strategy, particularly for patients with refractory or relapsed DLBCL. In this study, fatty-acid metabolism index (FMI) signature including 20 genes was constructed by univariate Cox and LASSO-Cox regression analysis using transcriptomic data from Gene Expression Omnibus database to predict overall survival of DLBCL patients. The FMI signature exhibited a negative association with anti-tumor immune response, with FMI-high patients had decreased immune cell infiltration and downregulated immune-associated signaling pathways. Based on the FMI signature, FASN was identified as the most essential gene that negatively regulated the tumor microenvironment of DLBCL. High FASN expression was associated with reduced anti-tumor immune activity, including decreased immune score, lower T-cell inflamed score, and downregulated expression of IFN molecules. Infiltration of immune cells, including CD4+T cells, CD8+T cells, dendritic cells, and macrophages were also significantly decreased in FASN-high than FASN-low patients. Accordingly, key signal molecules and chemokines for immune cells exhibited negative correlations with FASN expression. Through FASN knockdown in DB cell line, we further validated that FASN significantly suppressed chemokine secretion and promoted DLBCL proliferation in vitro. The FMI signature can effectively distinguish the prognostic stratification of DLBCL patients, further suggesting that FA metabolism imbalance may play an important role in DLBCL heterogeneity and treatment resistance. FASN is a potential negative regulator of immune microenvironment, providing novel insights into the metabolic-immune crosstalk in DLBCL treatment.

弥漫性大b细胞淋巴瘤(DLBCL)是最常见的非霍奇金淋巴瘤(NHL)亚型。靶向脂肪酸代谢途径是一种很有前景的治疗策略,特别是对于难治性或复发的DLBCL患者。本研究利用来自Gene Expression Omnibus数据库的转录组数据,通过单因素Cox和LASSO-Cox回归分析构建了包含20个基因的脂肪酸代谢指数(FMI)特征,预测DLBCL患者的总生存期。FMI特征与抗肿瘤免疫应答呈负相关,FMI高的患者免疫细胞浸润减少,免疫相关信号通路下调。基于FMI特征,FASN被确定为负向调控DLBCL肿瘤微环境的最重要基因。FASN高表达与抗肿瘤免疫活性降低相关,包括免疫评分降低、t细胞炎症评分降低和IFN分子表达下调。fasn高的患者CD4+T细胞、CD8+T细胞、树突状细胞、巨噬细胞等免疫细胞的浸润也明显低于fasn低的患者。因此,免疫细胞的关键信号分子和趋化因子与FASN表达呈负相关。通过在DB细胞系中敲低FASN,我们进一步验证了FASN在体外显著抑制趋化因子分泌,促进DLBCL增殖。FMI特征可以有效区分DLBCL患者的预后分层,进一步提示FA代谢失衡可能在DLBCL异质性和治疗耐药中起重要作用。FASN是免疫微环境的潜在负调节因子,为DLBCL治疗中的代谢-免疫串扰提供了新的见解。
{"title":"Fatty acid metabolic signature reveals FASN as an immunosuppressive factor in DLBCL tumor microenvironment.","authors":"Tingting Cheng, Shiyu Wang, Yue Zhang, Fenfang Tong, Wei Qin, Yajing Xu","doi":"10.1007/s12032-025-03159-5","DOIUrl":"https://doi.org/10.1007/s12032-025-03159-5","url":null,"abstract":"<p><p>Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin lymphoma (NHL). Targeting fatty acid metabolism pathway represents a promising therapeutic strategy, particularly for patients with refractory or relapsed DLBCL. In this study, fatty-acid metabolism index (FMI) signature including 20 genes was constructed by univariate Cox and LASSO-Cox regression analysis using transcriptomic data from Gene Expression Omnibus database to predict overall survival of DLBCL patients. The FMI signature exhibited a negative association with anti-tumor immune response, with FMI-high patients had decreased immune cell infiltration and downregulated immune-associated signaling pathways. Based on the FMI signature, FASN was identified as the most essential gene that negatively regulated the tumor microenvironment of DLBCL. High FASN expression was associated with reduced anti-tumor immune activity, including decreased immune score, lower T-cell inflamed score, and downregulated expression of IFN molecules. Infiltration of immune cells, including CD4<sup>+</sup>T cells, CD8<sup>+</sup>T cells, dendritic cells, and macrophages were also significantly decreased in FASN-high than FASN-low patients. Accordingly, key signal molecules and chemokines for immune cells exhibited negative correlations with FASN expression. Through FASN knockdown in DB cell line, we further validated that FASN significantly suppressed chemokine secretion and promoted DLBCL proliferation in vitro. The FMI signature can effectively distinguish the prognostic stratification of DLBCL patients, further suggesting that FA metabolism imbalance may play an important role in DLBCL heterogeneity and treatment resistance. FASN is a potential negative regulator of immune microenvironment, providing novel insights into the metabolic-immune crosstalk in DLBCL treatment.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"43 2","pages":"84"},"PeriodicalIF":3.5,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145834345","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
Icaritin suppresses CAD-mediated liver cancer development by targeting miR-18b-5p in a xenograft mouse model. 在异种移植小鼠模型中,icartin通过靶向miR-18b-5p抑制cad介导的肝癌发展。
IF 3.5 4区 医学 Q2 ONCOLOGY Pub Date : 2025-12-26 DOI: 10.1007/s12032-025-03211-4
Di Wu, Tian Mi, Xue Tang, Yiming Jia, Tao Guo, Guoqiang Zhou, Wenjuan Li

Cancer cells show abnormal nucleotide metabolism and prefer the de novo synthesis pathway. As the key enzymes, Carbamoyl-phosphate synthetase 2, Aspartate transcarbamoylase, and Dihydroorotase (CAD) is overactivated in cancer and promotes pyrimidine de novo synthesis, supplying cancer cells with DNA and RNA biosynthesis precursors. Therefore, the development of drugs targeting CAD might inhibit cancer progression and transformation. Icaritin (ICT) is an isoprenoid flavonoid derivative with a wide range of anticancer activities, however, the mechanism of ICT in regulating pyrimidine biosynthesis in cancer remains unclear. MicroRNAs are involved in carcinogenesis by regulating the expression of target genes, and ICT has been shown to regulate the expression of miRNAs leading to suppressing cancer progression. Using both human normal hepatocytes and liver cancer cells, we found that CAD expression was significantly elevated in cancer cells. Interestingly, although ICT treatment reduced CAD protein levels in liver cancer cells, it increased CAD transcriptional activity. Dual-luciferase reporter assays confirmed miR-18b-5p as a direct regulator of CAD. By transfecting miR-18b-5p mimics or inhibitors, we showed ICT upregulates miR-18b-5p to suppress CAD, inhibiting liver cancer cell proliferation, migration, and colony formation. Furthermore, in a human liver cancer xenograft mouse model, ICT treatment markedly reduced tumor growth and decreased Ki-67 expression, consistent with the in vitro results, CAD protein expression was downregulated, while its mRNA level was upregulated, further supporting a post-transcriptional regulatory mechanism. Overall, ICT plays an anti-liver cancer role by increasing miR-18b-5p at the post-transcriptional level to inhibit CAD expression, which may interfere with the de novo synthesis of pyrimidine and development of liver cancer.

癌细胞表现出异常的核苷酸代谢,并倾向于从头合成途径。作为关键酶,氨基甲酰磷酸合成酶2、天冬氨酸转氨基甲酰化酶和二氢羟化酶(CAD)在癌症中过度激活,促进嘧啶从头合成,为癌细胞提供DNA和RNA的生物合成前体。因此,开发针对CAD的药物可能会抑制癌症的进展和转化。icartin (icartin, ICT)是一类具有广泛抗癌活性的类黄酮类异戊二烯衍生物,但其在肿瘤中调节嘧啶生物合成的机制尚不清楚。microrna通过调节靶基因的表达参与癌变,而ICT已被证明可以调节microrna的表达从而抑制癌症的进展。在人类正常肝细胞和肝癌细胞中,我们发现CAD在癌细胞中的表达显著升高。有趣的是,尽管ICT治疗降低了肝癌细胞中的CAD蛋白水平,但却增加了CAD转录活性。双荧光素酶报告基因检测证实miR-18b-5p是CAD的直接调节因子。通过转染miR-18b-5p模拟物或抑制剂,我们发现ICT上调miR-18b-5p抑制CAD,抑制肝癌细胞增殖、迁移和集落形成。此外,在人肝癌异种移植小鼠模型中,ICT治疗显著抑制肿瘤生长,降低Ki-67表达,与体外实验结果一致,CAD蛋白表达下调,而其mRNA水平上调,进一步支持转录后调控机制。综上所述,ICT通过在转录后水平上增加miR-18b-5p抑制CAD的表达,从而起到抗肝癌的作用,这可能会干扰嘧啶的从头合成和肝癌的发展。
{"title":"Icaritin suppresses CAD-mediated liver cancer development by targeting miR-18b-5p in a xenograft mouse model.","authors":"Di Wu, Tian Mi, Xue Tang, Yiming Jia, Tao Guo, Guoqiang Zhou, Wenjuan Li","doi":"10.1007/s12032-025-03211-4","DOIUrl":"https://doi.org/10.1007/s12032-025-03211-4","url":null,"abstract":"<p><p>Cancer cells show abnormal nucleotide metabolism and prefer the de novo synthesis pathway. As the key enzymes, Carbamoyl-phosphate synthetase 2, Aspartate transcarbamoylase, and Dihydroorotase (CAD) is overactivated in cancer and promotes pyrimidine de novo synthesis, supplying cancer cells with DNA and RNA biosynthesis precursors. Therefore, the development of drugs targeting CAD might inhibit cancer progression and transformation. Icaritin (ICT) is an isoprenoid flavonoid derivative with a wide range of anticancer activities, however, the mechanism of ICT in regulating pyrimidine biosynthesis in cancer remains unclear. MicroRNAs are involved in carcinogenesis by regulating the expression of target genes, and ICT has been shown to regulate the expression of miRNAs leading to suppressing cancer progression. Using both human normal hepatocytes and liver cancer cells, we found that CAD expression was significantly elevated in cancer cells. Interestingly, although ICT treatment reduced CAD protein levels in liver cancer cells, it increased CAD transcriptional activity. Dual-luciferase reporter assays confirmed miR-18b-5p as a direct regulator of CAD. By transfecting miR-18b-5p mimics or inhibitors, we showed ICT upregulates miR-18b-5p to suppress CAD, inhibiting liver cancer cell proliferation, migration, and colony formation. Furthermore, in a human liver cancer xenograft mouse model, ICT treatment markedly reduced tumor growth and decreased Ki-67 expression, consistent with the in vitro results, CAD protein expression was downregulated, while its mRNA level was upregulated, further supporting a post-transcriptional regulatory mechanism. Overall, ICT plays an anti-liver cancer role by increasing miR-18b-5p at the post-transcriptional level to inhibit CAD expression, which may interfere with the de novo synthesis of pyrimidine and development of liver cancer.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"43 2","pages":"95"},"PeriodicalIF":3.5,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145834315","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
Role of Aldo-Keto reductase family 1 member C in cancer progression: a special focus on the role of AKR1C1 in pancreatic cancer. Aldo-Keto还原酶家族1成员C在癌症进展中的作用:AKR1C1在胰腺癌中的作用
IF 3.5 4区 医学 Q2 ONCOLOGY Pub Date : 2025-12-26 DOI: 10.1007/s12032-025-03234-x
Degao Huang, Heyang Zhang, Yu Zhang, Congliang Chen, Kexin Lin, Xia Wang, Peipei Wang

AKR1Cs, as a reductase enzyme family, play a pro-carcinogenic role in various types of cancers, including hormone-related malignancies and non-hormonal tumors. However, there exists a notable scarcity of literature concerning AKR1Cs expression in pancreatic cancer and the subsequent impacts on its progression. Analyzing pancreatic cancer database information by employing advanced bioinformatics techniques to unravel AKR1Cs' intricate involvement in cancer malignancy, their correlation with clinical pathology, prognostic implications, as well as their responsiveness to conventional and immune-based therapies. Furthermore, the role of AKR1C1 in promoting the malignant progression of pancreatic cancer cell lines was validated using cell proliferation assays (EdU labeling and colony formation), and cell migration and invasion experiments including scratch wound healing and Transwell migration/invasion assays. AKR1Cs are not only significantly overexpressed in pancreatic cancer, but also closely associated with poor clinical grading, clinical chemoresistance and poor immune response in pancreatic cancer.Moreover, regulating the expression of AKR1C1 in pancreatic cancer cells will affect its proliferation, migration, invasion and the occurrence of epithelial-mesenchymal transformation (EMT). Our findings are expected to establish AKR1Cs, especially AKR1C1 as a promising therapeutic target for the clinical treatment of pancreatic cancer.

akr1c作为一个还原酶家族,在各种类型的癌症中,包括激素相关的恶性肿瘤和非激素肿瘤中都起着促癌作用。然而,关于AKR1Cs在胰腺癌中的表达及其对其进展的影响的文献明显缺乏。通过采用先进的生物信息学技术分析胰腺癌数据库信息,揭示AKR1Cs在恶性肿瘤中的复杂参与,它们与临床病理、预后影响的相关性,以及它们对常规和免疫治疗的反应性。此外,通过细胞增殖实验(EdU标记和集落形成)、细胞迁移和侵袭实验(包括划伤愈合和Transwell迁移/侵袭实验)验证了AKR1C1在促进胰腺癌细胞系恶性进展中的作用。AKR1Cs不仅在胰腺癌中显著过表达,而且与胰腺癌临床分级差、临床化疗耐药和免疫应答差密切相关。此外,调节AKR1C1在胰腺癌细胞中的表达会影响其增殖、迁移、侵袭及上皮间充质转化(epithelial-mesenchymal transformation, EMT)的发生。我们的研究结果有望确立AKR1Cs,特别是AKR1C1作为胰腺癌临床治疗的有希望的治疗靶点。
{"title":"Role of Aldo-Keto reductase family 1 member C in cancer progression: a special focus on the role of AKR1C1 in pancreatic cancer.","authors":"Degao Huang, Heyang Zhang, Yu Zhang, Congliang Chen, Kexin Lin, Xia Wang, Peipei Wang","doi":"10.1007/s12032-025-03234-x","DOIUrl":"https://doi.org/10.1007/s12032-025-03234-x","url":null,"abstract":"<p><p>AKR1Cs, as a reductase enzyme family, play a pro-carcinogenic role in various types of cancers, including hormone-related malignancies and non-hormonal tumors. However, there exists a notable scarcity of literature concerning AKR1Cs expression in pancreatic cancer and the subsequent impacts on its progression. Analyzing pancreatic cancer database information by employing advanced bioinformatics techniques to unravel AKR1Cs' intricate involvement in cancer malignancy, their correlation with clinical pathology, prognostic implications, as well as their responsiveness to conventional and immune-based therapies. Furthermore, the role of AKR1C1 in promoting the malignant progression of pancreatic cancer cell lines was validated using cell proliferation assays (EdU labeling and colony formation), and cell migration and invasion experiments including scratch wound healing and Transwell migration/invasion assays. AKR1Cs are not only significantly overexpressed in pancreatic cancer, but also closely associated with poor clinical grading, clinical chemoresistance and poor immune response in pancreatic cancer.Moreover, regulating the expression of AKR1C1 in pancreatic cancer cells will affect its proliferation, migration, invasion and the occurrence of epithelial-mesenchymal transformation (EMT). Our findings are expected to establish AKR1Cs, especially AKR1C1 as a promising therapeutic target for the clinical treatment of pancreatic cancer.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"43 2","pages":"98"},"PeriodicalIF":3.5,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145834346","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
Understanding the role of vitamin D in osteosarcoma: A narrative review. 了解维生素D在骨肉瘤中的作用:一个叙述性的回顾。
IF 3.5 4区 医学 Q2 ONCOLOGY Pub Date : 2025-12-26 DOI: 10.1007/s12032-025-03162-w
Dian Caturini Sulistyoningrum, Yuni Artha Prabowo Putro, Annisa Fitria Nur Azizah, Amri Wicaksono Pribadi, Ery Kus Dwianingsih

This review provides a summary of recent evidence on vitamin D and osteosarcoma, aiming to elucidate the molecular mechanisms of vitamin D in osteosarcoma and explore its potential role in patient management. Osteosarcoma, a malignant bone neoplasm, is characterized by its aggressive nature, poor prognosis, and metastatic potential, often spreading to the lungs. Its clinical severity highlights the need for early detection, prompt intervention, and comprehensive treatment strategies to improve outcomes and quality of life for individuals affected by it. Circulating 25-hydroxyvitamin D [25(OH)D] is quantified as a measure of vitamin D status. Vitamin D status is utilized as a clinical biomarker to identify vitamin D deficiency. The role of vitamin D in bone health and development is well known, specifically its regulation of calcium-phosphate homeostasis. Furthermore, recent findings identified vitamin D's role in carcinogenesis by regulating cancer cell differentiation, proliferation, apoptosis, and metastatic potential. The involvement of vitamin D-binding protein (VDBP) and its polymorphisms in regulating vitamin D bioavailability has also been recognized. However, these findings are primarily in major cancers such as breast cancer, lung cancer, and colorectal cancer. Evidence pertaining to the fundamental role of bone cancer, in particular osteosarcoma, and vitamin D remains scarce. Nevertheless, the limited evidence showed that maintaining circulating 25(OH)D in osteosarcoma patients is associated with a higher survival rate. Meanwhile, vitamin D supplementation as an adjuvant could potentially increase survival rates and quality of life.

本文综述了近年来有关维生素D与骨肉瘤的研究进展,旨在阐明维生素D在骨肉瘤中的分子机制,并探讨其在患者治疗中的潜在作用。骨肉瘤是一种恶性骨肿瘤,其特点是具有侵袭性、预后差和转移潜力,常扩散到肺部。其临床严重性突出了早期发现、及时干预和综合治疗策略的必要性,以改善受其影响的个体的预后和生活质量。循环25-羟基维生素D [25(OH)D]被量化为维生素D状态的测量。维生素D状态被用作确定维生素D缺乏的临床生物标志物。维生素D在骨骼健康和发育中的作用是众所周知的,特别是它对磷酸钙稳态的调节。此外,最近的研究发现,维生素D通过调节癌细胞分化、增殖、凋亡和转移潜能,在致癌过程中发挥作用。维生素D结合蛋白(VDBP)及其多态性在调节维生素D生物利用度中的作用也已得到确认。然而,这些发现主要是针对乳腺癌、肺癌和结直肠癌等主要癌症。有关骨癌,特别是骨肉瘤和维生素D的基本作用的证据仍然很少。然而,有限的证据表明,维持骨肉瘤患者循环25(OH)D与更高的生存率相关。同时,补充维生素D作为辅助剂可能会潜在地提高生存率和生活质量。
{"title":"Understanding the role of vitamin D in osteosarcoma: A narrative review.","authors":"Dian Caturini Sulistyoningrum, Yuni Artha Prabowo Putro, Annisa Fitria Nur Azizah, Amri Wicaksono Pribadi, Ery Kus Dwianingsih","doi":"10.1007/s12032-025-03162-w","DOIUrl":"https://doi.org/10.1007/s12032-025-03162-w","url":null,"abstract":"<p><p>This review provides a summary of recent evidence on vitamin D and osteosarcoma, aiming to elucidate the molecular mechanisms of vitamin D in osteosarcoma and explore its potential role in patient management. Osteosarcoma, a malignant bone neoplasm, is characterized by its aggressive nature, poor prognosis, and metastatic potential, often spreading to the lungs. Its clinical severity highlights the need for early detection, prompt intervention, and comprehensive treatment strategies to improve outcomes and quality of life for individuals affected by it. Circulating 25-hydroxyvitamin D [25(OH)D] is quantified as a measure of vitamin D status. Vitamin D status is utilized as a clinical biomarker to identify vitamin D deficiency. The role of vitamin D in bone health and development is well known, specifically its regulation of calcium-phosphate homeostasis. Furthermore, recent findings identified vitamin D's role in carcinogenesis by regulating cancer cell differentiation, proliferation, apoptosis, and metastatic potential. The involvement of vitamin D-binding protein (VDBP) and its polymorphisms in regulating vitamin D bioavailability has also been recognized. However, these findings are primarily in major cancers such as breast cancer, lung cancer, and colorectal cancer. Evidence pertaining to the fundamental role of bone cancer, in particular osteosarcoma, and vitamin D remains scarce. Nevertheless, the limited evidence showed that maintaining circulating 25(OH)D in osteosarcoma patients is associated with a higher survival rate. Meanwhile, vitamin D supplementation as an adjuvant could potentially increase survival rates and quality of life.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"43 2","pages":"74"},"PeriodicalIF":3.5,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145834349","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
期刊
Medical Oncology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
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
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1