首页 > 最新文献

Oncology reports最新文献

英文 中文
Evaluation of co‑inhibition of ErbB family kinases and PI3K for HPV‑negative head and neck squamous cell carcinoma.
IF 3.8 3区 医学 Q2 ONCOLOGY Pub Date : 2025-03-01 Epub Date: 2025-01-31 DOI: 10.3892/or.2025.8871
Xinyan Geng, Shirin Azarbarzin, Zejia Yang, Rena G Lapidus, Xiaoxuan Fan, Yong Teng, Ranee Mehra, Kevin J Cullen, Hancai Dan

The ErbB/HER family of protein‑tyrosine kinases and PI3K represent crucial targets in the treatment of head and neck squamous cell carcinoma (HNSCC). A combination therapy of afatinib (ErbB inhibitor) and copanlisib (PI3K inhibitor), both Food and Drug Administration‑approved kinase inhibitors, can suppress the growth of human papillomavirus (HPV)‑positive HNSCC. The current study further evaluated the efficacy and clinical potential of this combination therapy for the treatment of HPV‑negative HNSCC in vitro and in vivo. Sulforhodamine B cell viability assay and Annexin V/propidium iodide staining demonstrated that this combination treatment markedly enhanced inhibition of cell viability and reduced cell survival when compared with treatment with either inhibitor alone in two HPV‑negative HNSCC cell lines. Notably, this combination also led to significant inhibition of xenograft tumor growth in mice, without any apparent effects on body weight. Western blot analysis found that copanlisib alone effectively blocked PI3K/Akt signaling but caused upregulation of HER2 and HER3 phosphorylation, as reported in other types of cancer. However, the combination of copanlisib and afatinib completely blocked phosphorylation of the ErbB family (including HER3) and Akt, while also increasing apoptosis. In conclusion, these results suggested that co‑targeting the ErbB family kinases and PI3K using a combination treatment of afatinib and copanlisib may have clinical potential for patients with HPV‑negative HNSCC.

{"title":"Evaluation of co‑inhibition of ErbB family kinases and PI3K for HPV‑negative head and neck squamous cell carcinoma.","authors":"Xinyan Geng, Shirin Azarbarzin, Zejia Yang, Rena G Lapidus, Xiaoxuan Fan, Yong Teng, Ranee Mehra, Kevin J Cullen, Hancai Dan","doi":"10.3892/or.2025.8871","DOIUrl":"https://doi.org/10.3892/or.2025.8871","url":null,"abstract":"<p><p>The ErbB/HER family of protein‑tyrosine kinases and PI3K represent crucial targets in the treatment of head and neck squamous cell carcinoma (HNSCC). A combination therapy of afatinib (ErbB inhibitor) and copanlisib (PI3K inhibitor), both Food and Drug Administration‑approved kinase inhibitors, can suppress the growth of human papillomavirus (HPV)‑positive HNSCC. The current study further evaluated the efficacy and clinical potential of this combination therapy for the treatment of HPV‑negative HNSCC <i>in vitro</i> and <i>in vivo</i>. Sulforhodamine B cell viability assay and Annexin V/propidium iodide staining demonstrated that this combination treatment markedly enhanced inhibition of cell viability and reduced cell survival when compared with treatment with either inhibitor alone in two HPV‑negative HNSCC cell lines. Notably, this combination also led to significant inhibition of xenograft tumor growth in mice, without any apparent effects on body weight. Western blot analysis found that copanlisib alone effectively blocked PI3K/Akt signaling but caused upregulation of HER2 and HER3 phosphorylation, as reported in other types of cancer. However, the combination of copanlisib and afatinib completely blocked phosphorylation of the ErbB family (including HER3) and Akt, while also increasing apoptosis. In conclusion, these results suggested that co‑targeting the ErbB family kinases and PI3K using a combination treatment of afatinib and copanlisib may have clinical potential for patients with HPV‑negative HNSCC.</p>","PeriodicalId":19527,"journal":{"name":"Oncology reports","volume":"53 3","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143066272","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Triple‑negative breast cancer cell‑derived piR‑31115 promotes the proliferation and migration of endothelial cells via METTL3‑mediated m6A modification of YAP1. 三阴性乳腺癌细胞源性piR - 31115通过METTL3介导的m6A修饰YAP1促进内皮细胞的增殖和迁移。
IF 3.8 3区 医学 Q2 ONCOLOGY Pub Date : 2025-03-01 Epub Date: 2025-01-17 DOI: 10.3892/or.2025.8867
Shan-Mei Du, Na Li, Wen-Jing Xu, Kui Liu

Triple‑negative breast cancer (TNBC), a highly malignant breast cancer subtype with a pronounced metastatic propensity, forms the focus of the present investigation. MDA‑MB‑231, a prevalently utilized TNBC cell line in cancer research, was employed. In accordance with the tumour angiogenesis theory, cancer cells are capable of instigating angiogenesis and the formation of a novel vascular system within the tumour microenvironment, which subsequently sustains malignant proliferation and metastasis. Consequently, impeding the growth of tumour blood vessels holds substantial significance in suppressing TNBC metastasis. Piwi‑interacting RNAs (piRNAs), a category of endogenous non‑coding RNAs, have been demonstrated to modulate cancer progression. However, studies regarding the role of piRNAs in regulating angiogenesis within cancer cells are relatively scant. In the present study, via cell co‑culture experiments, it was revealed that piR‑31115 (a kind of piRNA) in MDA‑MB‑231 cells notably enhanced the recruitment of a human microvascular endothelial cell line (HMEC‑1). Moreover, the conditioned medium (CM, which was obtained from MDA‑MB‑231 cells via a specific culturing methodology and was employed for the subsequent treatment of HMEC‑1 cells to explore its impacts on the biological behaviors such as the proliferation and migration of HMEC‑1 cells) derived from MDA‑MB‑231 cells with upregulated piR‑31115 expression stimulated the proliferation and migration of HMEC‑1 cells. These findings suggest that piR‑31115 in MDA‑MB‑231 cells may play a pivotal role in modulating tumour angiogenesis. Further studies disclosed that the CM from MDA‑MB‑231 cells augmented the N6‑methyladenosine (m6A) RNA modification level via METTL3 in HMEC‑1 cells. Transcriptome sequencing revealed that METTL3 functions as an m6A writer protein for Yes‑associated protein 1 (YAP1), which exerts a positive influence on promoting the proliferation and migration of HMEC‑1 cells. Concurrently, the IGF2BP plays a crucial role in stabilizing YAP1 protein expression. Collectively, the present findings identified a signalling pathway through which MDA‑MB‑231 cells induce HMEC‑1 cell proliferation and migration by regulating m6A RNA methylation.

三阴性乳腺癌(TNBC)是一种高度恶性的乳腺癌亚型,具有明显的转移倾向,是本研究的重点。MDA - MB - 231是癌症研究中普遍使用的TNBC细胞系。根据肿瘤血管生成理论,癌细胞能够在肿瘤微环境中诱导血管生成,形成新的血管系统,从而维持恶性肿瘤的增殖和转移。因此,抑制肿瘤血管生长对抑制TNBC转移具有重要意义。Piwi相互作用rna (piRNAs)是一类内源性非编码rna,已被证明可以调节癌症的进展。然而,关于pirna在调节癌细胞血管生成中的作用的研究相对较少。在本研究中,通过细胞共培养实验,发现MDA‑MB‑231细胞中的piR‑31115(一种piRNA)显著增强了人微血管内皮细胞系(HMEC‑1)的募集。此外,piR - 31115表达上调的MDA - MB - 231细胞衍生的条件培养基(CM,通过特殊培养方法从MDA - MB - 231细胞中获得,用于HMEC - 1细胞的后续处理,以探索其对HMEC - 1细胞增殖和迁移等生物学行为的影响)刺激了HMEC - 1细胞的增殖和迁移。这些发现表明MDA - MB - 231细胞中的piR - 31115可能在调节肿瘤血管生成中起关键作用。进一步的研究表明,MDA - MB - 231细胞中的CM通过METTL3增强了HMEC - 1细胞中N6 -甲基腺苷(m6A) RNA修饰水平。转录组测序结果显示,METTL3作为Yes - associated protein 1 (YAP1)的m6A写蛋白,对促进HMEC - 1细胞的增殖和迁移具有积极作用。同时,IGF2BP在稳定YAP1蛋白表达中起着至关重要的作用。总的来说,目前的研究结果确定了MDA‑MB‑231细胞通过调节m6A RNA甲基化诱导HMEC‑1细胞增殖和迁移的信号通路。
{"title":"Triple‑negative breast cancer cell‑derived piR‑31115 promotes the proliferation and migration of endothelial cells via METTL3‑mediated m6A modification of YAP1.","authors":"Shan-Mei Du, Na Li, Wen-Jing Xu, Kui Liu","doi":"10.3892/or.2025.8867","DOIUrl":"10.3892/or.2025.8867","url":null,"abstract":"<p><p>Triple‑negative breast cancer (TNBC), a highly malignant breast cancer subtype with a pronounced metastatic propensity, forms the focus of the present investigation. MDA‑MB‑231, a prevalently utilized TNBC cell line in cancer research, was employed. In accordance with the tumour angiogenesis theory, cancer cells are capable of instigating angiogenesis and the formation of a novel vascular system within the tumour microenvironment, which subsequently sustains malignant proliferation and metastasis. Consequently, impeding the growth of tumour blood vessels holds substantial significance in suppressing TNBC metastasis. Piwi‑interacting RNAs (piRNAs), a category of endogenous non‑coding RNAs, have been demonstrated to modulate cancer progression. However, studies regarding the role of piRNAs in regulating angiogenesis within cancer cells are relatively scant. In the present study, via cell co‑culture experiments, it was revealed that piR‑31115 (a kind of piRNA) in MDA‑MB‑231 cells notably enhanced the recruitment of a human microvascular endothelial cell line (HMEC‑1). Moreover, the conditioned medium (CM, which was obtained from MDA‑MB‑231 cells via a specific culturing methodology and was employed for the subsequent treatment of HMEC‑1 cells to explore its impacts on the biological behaviors such as the proliferation and migration of HMEC‑1 cells) derived from MDA‑MB‑231 cells with upregulated piR‑31115 expression stimulated the proliferation and migration of HMEC‑1 cells. These findings suggest that piR‑31115 in MDA‑MB‑231 cells may play a pivotal role in modulating tumour angiogenesis. Further studies disclosed that the CM from MDA‑MB‑231 cells augmented the N6‑methyladenosine (m6A) RNA modification level via METTL3 in HMEC‑1 cells. Transcriptome sequencing revealed that METTL3 functions as an m6A writer protein for Yes‑associated protein 1 (YAP1), which exerts a positive influence on promoting the proliferation and migration of HMEC‑1 cells. Concurrently, the IGF2BP plays a crucial role in stabilizing YAP1 protein expression. Collectively, the present findings identified a signalling pathway through which MDA‑MB‑231 cells induce HMEC‑1 cell proliferation and migration by regulating m6A RNA methylation.</p>","PeriodicalId":19527,"journal":{"name":"Oncology reports","volume":"53 3","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11755246/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143009124","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
[Retracted] Inhibition of neddylation modification by MLN4924 sensitizes hepatocellular carcinoma cells to sorafenib. 【撤回】MLN4924抑制类化修饰可使肝癌细胞对索拉非尼增敏。
IF 3.8 3区 医学 Q2 ONCOLOGY Pub Date : 2025-03-01 Epub Date: 2025-01-17 DOI: 10.3892/or.2025.8866
Zelong Yang, Jie Zhang, Xiaotong Lin, Di Wu, Guixi Li, Chunlian Zhong, Lei Fang, Peng Jiang, Liangyu Yin, Leida Zhang, Ping Bie, Chuan-Ming Xie

Following the publication of the above article, the authors noticed that they had inadvertently included a duplication of the same data in Fig. 3C, portraying colony formation experiments, where the results from differently performed experiments were intended to have been shown, and requested that a corrigendum be published to present the data in this figure accurately. Having investigated this matter in the Editorial Office, however, additional panels of overlapping data were identified, comparing between Figs. 2 and 3; moreover, a pair of overlapping data panels were also identified examining the Transwell migration assay data in Fig. 5A. The Editor of Oncology Reports has considered the authors' request to publish a corrigendum, but has decided to decline this request on account of the additional errors that have been identified; rather, the article is to be be retracted from the Journal on the basis of an overall lack of confidence in the presented data. Upon receiving this decision from the Editor, the authors did not provide a satisfactory reply. The Editor apologizes to the readership of the Journal for any inconvenience caused.  [Oncology Reports 41: 3257‑3269, 2019; DOI: 10.3892/or.2019.7098].

在上述文章发表后,作者注意到他们无意中在图3C中包含了相同数据的重复,图3C描绘了菌落形成实验,其中本该显示不同实验的结果,并要求发布更正以准确呈现该图中的数据。然而,在编辑部调查了这个问题后,发现了额外的重叠数据面板,比较图2和图3;此外,在图5A中还发现了一对重叠的数据面板,用于检查Transwell迁移分析数据。《肿瘤学报告》编辑考虑了作者发表更正的请求,但由于发现了额外的错误,决定拒绝这一请求;相反,这篇文章将从《华尔街日报》上撤回,理由是对所提供的数据总体上缺乏信心。在收到编辑的这一决定后,作者没有提供令人满意的答复。编辑为给《华尔街日报》读者带来的不便向读者道歉。[肿瘤杂志]41:3257‑3269,2019;DOI: 10.3892 / or.2019.7098]。
{"title":"[Retracted] Inhibition of neddylation modification by MLN4924 sensitizes hepatocellular carcinoma cells to sorafenib.","authors":"Zelong Yang, Jie Zhang, Xiaotong Lin, Di Wu, Guixi Li, Chunlian Zhong, Lei Fang, Peng Jiang, Liangyu Yin, Leida Zhang, Ping Bie, Chuan-Ming Xie","doi":"10.3892/or.2025.8866","DOIUrl":"10.3892/or.2025.8866","url":null,"abstract":"<p><p>Following the publication of the above article, the authors noticed that they had inadvertently included a duplication of the same data in Fig. 3C, portraying colony formation experiments, where the results from differently performed experiments were intended to have been shown, and requested that a corrigendum be published to present the data in this figure accurately. Having investigated this matter in the Editorial Office, however, additional panels of overlapping data were identified, comparing between Figs. 2 and 3; moreover, a pair of overlapping data panels were also identified examining the Transwell migration assay data in Fig. 5A. The Editor of <i>Oncology Reports</i> has considered the authors' request to publish a corrigendum, but has decided to decline this request on account of the additional errors that have been identified; rather, the article is to be be retracted from the Journal on the basis of an overall lack of confidence in the presented data. Upon receiving this decision from the Editor, the authors did not provide a satisfactory reply. The Editor apologizes to the readership of the Journal for any inconvenience caused.  [Oncology Reports 41: 3257‑3269, 2019; DOI: 10.3892/or.2019.7098].</p>","PeriodicalId":19527,"journal":{"name":"Oncology reports","volume":"53 3","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11747261/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143009122","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Role of miRNA‑145‑5p in cancer (Review).
IF 3.8 3区 医学 Q2 ONCOLOGY Pub Date : 2025-03-01 Epub Date: 2025-01-31 DOI: 10.3892/or.2025.8872
Zeshan Chen, Yijue Qin

MicroRNA‑145‑5p (miRNA‑145‑5p) is a short non‑coding RNA located at chromosome 5q33.1, which has gained significant attention in several aspects of cellular regulation and biological functions. In malignant tumours, miRNA‑145‑5p may function as either a tumour suppressor or an oncogene, affecting tumour progression by targeting downstream genes or modulating their expression through upstream regulators. However, the full extent of miRNA‑145‑5p's role in cancer has remained to be determined. This review provides an overview of the role of miRNA‑145‑5p in cancer, investigates its potential as a biomarker for diagnosis, prognosis and treatment response, and evaluates its influence on cancer chemotherapy and radiotherapy. Finally, current strategies for systemic delivery of miRNA‑145‑5p in cancer therapies are summarized.

{"title":"Role of miRNA‑145‑5p in cancer (Review).","authors":"Zeshan Chen, Yijue Qin","doi":"10.3892/or.2025.8872","DOIUrl":"https://doi.org/10.3892/or.2025.8872","url":null,"abstract":"<p><p>MicroRNA‑145‑5p (miRNA‑145‑5p) is a short non‑coding RNA located at chromosome 5q33.1, which has gained significant attention in several aspects of cellular regulation and biological functions. In malignant tumours, miRNA‑145‑5p may function as either a tumour suppressor or an oncogene, affecting tumour progression by targeting downstream genes or modulating their expression through upstream regulators. However, the full extent of miRNA‑145‑5p's role in cancer has remained to be determined. This review provides an overview of the role of miRNA‑145‑5p in cancer, investigates its potential as a biomarker for diagnosis, prognosis and treatment response, and evaluates its influence on cancer chemotherapy and radiotherapy. Finally, current strategies for systemic delivery of miRNA‑145‑5p in cancer therapies are summarized.</p>","PeriodicalId":19527,"journal":{"name":"Oncology reports","volume":"53 3","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143066471","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Heteronemin suppresses EGF‑induced proliferation through the PI3K/PD‑L1 signaling pathways in cholangiocarcinoma. 在胆管癌中,Heteronemin通过PI3K/PD - L1信号通路抑制EGF诱导的增殖。
IF 3.8 3区 医学 Q2 ONCOLOGY Pub Date : 2025-03-01 Epub Date: 2025-01-10 DOI: 10.3892/or.2025.8865
Yu-Chen S H Yang, Chung-Che Tsai, Yung-Ning Yang, Feng-Cheng Liu, Sheng-Yang Lee, Jen-Chang Yang, Dana R Crawford, Hsien-Chung Chiu, Mei-Chin Lu, Zi-Lin Li, Yi-Chen Chen, Tin-Yi Chu, Jacqueline Whang-Peng, Hung-Yun Lin, Kuan Wang

Epidermal growth factor (EGF) binds with its surface receptor to stimulate gene expression and cancer cell proliferation. EGF stimulates cancer cell growth via phosphoinositide 3‑kinase (PI3K) and programmed cell death ligand 1 (PD‑L1) pathways. As an integrin αvβ3 antagonist, heteronemin exhibits potent cytotoxic effects against cancer cells. It inhibits critical signal transduction pathways promoted by the EGF. In the current study, EGF‑induced signal activation and proliferative effects were investigated in cholangiocarcinoma cells and its molecular targets using qPCR and western blotting analyses. In addition, cell viability assays were performed to assess the growth effects of EGF and heteronemin. Heteronemin reversed the effects of EGF and was further enhanced by blockage of PI3K's activity. In summary, EGF stimulates cholangiocarcinoma cell growth. On the other hand, heteronemin inhibited PI3K activation and PD‑L1 expression to reverse the stimulative effects of EGF‑induced gene expression and proliferation in cholangiocarcinoma cells.

表皮生长因子(EGF)与其表面受体结合,刺激基因表达和癌细胞增殖。EGF通过磷酸肌肽3激酶(PI3K)和程序性细胞死亡配体1 (PD - L1)途径刺激癌细胞生长。作为一种整合素αvβ3拮抗剂,异酮胺对癌细胞具有很强的细胞毒作用。它抑制EGF促进的关键信号转导途径。本研究采用qPCR和western blotting分析方法,研究了EGF诱导的信号激活及其在胆管癌细胞及其分子靶点中的增殖作用。此外,还进行了细胞活力测定,以评估EGF和异黄酮对细胞生长的影响。Heteronemin逆转了EGF的作用,并通过阻断PI3K的活性进一步增强。总之,EGF刺激胆管癌细胞生长。另一方面,heteronemin抑制PI3K活化和PD - L1表达,逆转EGF诱导的胆管癌细胞基因表达和增殖的刺激作用。
{"title":"Heteronemin suppresses EGF‑induced proliferation through the PI3K/PD‑L1 signaling pathways in cholangiocarcinoma.","authors":"Yu-Chen S H Yang, Chung-Che Tsai, Yung-Ning Yang, Feng-Cheng Liu, Sheng-Yang Lee, Jen-Chang Yang, Dana R Crawford, Hsien-Chung Chiu, Mei-Chin Lu, Zi-Lin Li, Yi-Chen Chen, Tin-Yi Chu, Jacqueline Whang-Peng, Hung-Yun Lin, Kuan Wang","doi":"10.3892/or.2025.8865","DOIUrl":"10.3892/or.2025.8865","url":null,"abstract":"<p><p>Epidermal growth factor (EGF) binds with its surface receptor to stimulate gene expression and cancer cell proliferation. EGF stimulates cancer cell growth via phosphoinositide 3‑kinase (PI3K) and programmed cell death ligand 1 (PD‑L1) pathways. As an integrin αvβ3 antagonist, heteronemin exhibits potent cytotoxic effects against cancer cells. It inhibits critical signal transduction pathways promoted by the EGF. In the current study, EGF‑induced signal activation and proliferative effects were investigated in cholangiocarcinoma cells and its molecular targets using qPCR and western blotting analyses. In addition, cell viability assays were performed to assess the growth effects of EGF and heteronemin. Heteronemin reversed the effects of EGF and was further enhanced by blockage of PI3K's activity. In summary, EGF stimulates cholangiocarcinoma cell growth. On the other hand, heteronemin inhibited PI3K activation and PD‑L1 expression to reverse the stimulative effects of EGF‑induced gene expression and proliferation in cholangiocarcinoma cells.</p>","PeriodicalId":19527,"journal":{"name":"Oncology reports","volume":"53 3","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142952657","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
[Corrigendum] ND‑09 inhibits chronic myeloid leukemia K562 cell growth by regulating BCR‑ABL signaling.
IF 3.8 3区 医学 Q2 ONCOLOGY Pub Date : 2025-03-01 Epub Date: 2025-01-31 DOI: 10.3892/or.2025.8870
Yan-Hong Liu, Man Zhu, Pan-Pan Lei, Xiao-Yan Pan, Wei-Na Ma

Subsequently to the publication of the above article, and a Corrigendum that has already been published with the intention of showing a corrected version of Fig. 2A (DOI: 10.3892/or.2023.8518; published online on March 3, 2023), the authors have subsequently realized that other errors were featured in certain of the published figures. First, the authors realized that Fig. 3 on p. 5 was incorrectly assembled: specifically, the flow cytometric data included in Figs. 3A and 3B were inadvertently assembled incorrectly. Additionally, the authors note that the control GAPDH data were duplicated in Figs. 5A and 5B, 6A and 6B, and 7A and 7B on p. 7 and 8. These errors occurred on account of the fact that the authors chose to use the same GAPDH bands for quantitative analysis, which they subsequently realize was not an appropriate course of action; the original GAPDH bands pertaining to the correct experiments are now included in each of these figures. The revised versions of Figs. 3, 5, 6 and 7 are shown on the next two pages. Note that the revisions made to Figs. 3, 5, 6 and 7 in this paper do not have a major impact on the reported results, and neither do they affect the overall conclusions reported in the study. All the authors agree to the publication of this corrigendum. The authors are grateful to the Editor of Oncology Reports for allowing them the opportunity to publish this additional Corrigendum; furthermore, they apologize for any inconvenience caused to the readership of the Journal. [Oncology Reports 46: 136, 2021; DOI: 10.3892/or.2021.8087].

{"title":"[Corrigendum] ND‑09 inhibits chronic myeloid leukemia K562 cell growth by regulating BCR‑ABL signaling.","authors":"Yan-Hong Liu, Man Zhu, Pan-Pan Lei, Xiao-Yan Pan, Wei-Na Ma","doi":"10.3892/or.2025.8870","DOIUrl":"https://doi.org/10.3892/or.2025.8870","url":null,"abstract":"<p><p>Subsequently to the publication of the above article, and a Corrigendum that has already been published with the intention of showing a corrected version of Fig. 2A (DOI: 10.3892/or.2023.8518; published online on March 3, 2023), the authors have subsequently realized that other errors were featured in certain of the published figures. First, the authors realized that Fig. 3 on p. 5 was incorrectly assembled: specifically, the flow cytometric data included in Figs. 3A and 3B were inadvertently assembled incorrectly. Additionally, the authors note that the control GAPDH data were duplicated in Figs. 5A and 5B, 6A and 6B, and 7A and 7B on p. 7 and 8. These errors occurred on account of the fact that the authors chose to use the same GAPDH bands for quantitative analysis, which they subsequently realize was not an appropriate course of action; the original GAPDH bands pertaining to the correct experiments are now included in each of these figures. The revised versions of Figs. 3, 5, 6 and 7 are shown on the next two pages. Note that the revisions made to Figs. 3, 5, 6 and 7 in this paper do not have a major impact on the reported results, and neither do they affect the overall conclusions reported in the study. All the authors agree to the publication of this corrigendum. The authors are grateful to the Editor of <i>Oncology Reports</i> for allowing them the opportunity to publish this additional Corrigendum; furthermore, they apologize for any inconvenience caused to the readership of the Journal. [Oncology Reports 46: 136, 2021; DOI: 10.3892/or.2021.8087].</p>","PeriodicalId":19527,"journal":{"name":"Oncology reports","volume":"53 3","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143067232","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
[Retracted] Inhibition of 5‑lipoxygenase triggers apoptosis in pancreatic cancer cells. [撤稿】抑制 5-脂氧合酶可引发胰腺癌细胞凋亡。
IF 3.8 3区 医学 Q2 ONCOLOGY Pub Date : 2025-02-01 Epub Date: 2024-12-13 DOI: 10.3892/or.2024.8853
Guo-Xiong Zhou, Xiao-Ling Ding, Sheng-Bao Wu, Hai-Feng Zhang, Wei Cao, Li-Shuai Qu, Hong Zhang

Following the publication of the above paper, a concerned reader drew to the Editor's attention that Fig. 3 on p. 664, showing TUNEL assay data relating to apoptosis of the cell line under investigation in this paper, contained apparent anomalies, including repeated patternings of certain cells both within and between the data panels, such that it would have been difficult to have attributed these anomalies to coincidence. After having conducted an independent investigation in the Editorial Office, the Editor of Oncology Reports has determined that the above paper should be retracted from the Journal on account of a lack of confidence concerning the originality and the authenticity of the data. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a satisfactory reply. The Editor regrets any inconvenience that has been caused to the readership of the Journal. [Oncology Reports 33: 661‑668, 2015; DOI: 10.3892/or.2014.3650].

在上述论文发表后,一位相关读者提请编辑注意,第 664 页的图 3 显示了与本文研究的细胞系凋亡有关的 TUNEL 检测数据,其中包含明显的异常现象,包括某些细胞在数据板内和数据板之间的重复图案,因此很难将这些异常现象归因于巧合。在编辑部进行独立调查后,《肿瘤学报告》编辑认为,由于对数据的原创性和真实性缺乏信心,上述论文应从杂志上撤下。编辑部要求作者就这些问题做出解释,但未收到令人满意的答复。对于给本刊读者造成的不便,编辑深表遗憾。[肿瘤学报告 33: 661-668, 2015; DOI: 10.3892/or.2014.3650]。
{"title":"[Retracted] Inhibition of 5‑lipoxygenase triggers apoptosis in pancreatic cancer cells.","authors":"Guo-Xiong Zhou, Xiao-Ling Ding, Sheng-Bao Wu, Hai-Feng Zhang, Wei Cao, Li-Shuai Qu, Hong Zhang","doi":"10.3892/or.2024.8853","DOIUrl":"10.3892/or.2024.8853","url":null,"abstract":"<p><p>Following the publication of the above paper, a concerned reader drew to the Editor's attention that Fig. 3 on p. 664, showing TUNEL assay data relating to apoptosis of the cell line under investigation in this paper, contained apparent anomalies, including repeated patternings of certain cells both within and between the data panels, such that it would have been difficult to have attributed these anomalies to coincidence. After having conducted an independent investigation in the Editorial Office, the Editor of <i>Oncology Reports</i> has determined that the above paper should be retracted from the Journal on account of a lack of confidence concerning the originality and the authenticity of the data. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a satisfactory reply. The Editor regrets any inconvenience that has been caused to the readership of the Journal. [Oncology Reports 33: 661‑668, 2015; DOI: 10.3892/or.2014.3650].</p>","PeriodicalId":19527,"journal":{"name":"Oncology reports","volume":"53 2","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11652959/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142818627","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Regulation of pancreatic cancer cells by suppressing KIN17 through the PI3K/AKT/mTOR signaling pathway. 通过PI3K/AKT/mTOR信号通路抑制KIN17调控胰腺癌细胞。
IF 3.8 3区 医学 Q2 ONCOLOGY Pub Date : 2025-02-01 Epub Date: 2025-01-10 DOI: 10.3892/or.2025.8864
Qiuyan Li, Yuxia Yang, Xiaocong Lin, Lok Ting Chu, Helian Chen, Linsong Chen, Jinjing Tang, Tao Zeng

Pancreatic cancer is an aggressive tumor, which is often associated with a poor clinical prognosis and resistance to conventional chemotherapy. Therefore, there is a need to identify new therapeutic markers for pancreatic cancer. Although KIN17 is a highly expressed DNA‑ and RNA‑binding protein in a number of types of human cancer, its role in pancreatic cancer development, especially in relation to progression, is currently unknown. The present study verified the upregulation of KIN17 in pancreatic cancer using The Cancer Genome Atlas and Gene Expression Omnibus databases (GSE15471, GSE71989 and GSE62165), and identified an association between the PI3K/Akt/mTOR pathway and patient prognosis using publicly available datasets (Gene Expression Profiling Interactive Analysis). Immunohistochemistry was performed to determine the association between KIN17 and the pathological features of clinical pancreatic cancer samples. Furthermore, knockdown of KIN17 was shown to inhibit the migration and invasion of pancreatic cancer cells, and to reverse epithelial‑mesenchymal transition in pancreatic cancer cells through downregulation of Vimentin and N‑cadherin, and upregulation of E‑cadherin. Through various cellular experiments, the role of KIIN17 was explored in PI3K/AKT/mTOR activity. KIN17 inhibition was shown to suppress the migration and invasion of pancreatic cancer cells through PI3K/AKT/mTOR‑mediated autophagy. Furthermore, combined with mTOR inhibition, dual inhibition could enhance autophagy, leading to anti‑migratory and anti‑invasion effects in pancreatic cancer. In conclusion, the present study indicated that KIN17 may have a role in carcinogenesis and could serve as a prognostic biomarker of pancreatic cancer, owing to its high expression. In addition, KIN17 may be considered a potential therapeutic target with its knockdown having an inhibitory effect on pancreatic cancer.

胰腺癌是一种侵袭性肿瘤,通常与临床预后差和常规化疗耐药有关。因此,有必要寻找新的胰腺癌治疗标志物。尽管KIN17在许多人类癌症中是一种高表达的DNA和RNA结合蛋白,但其在胰腺癌发展中的作用,特别是与进展的关系,目前尚不清楚。本研究利用cancer Genome Atlas和Gene Expression Omnibus数据库(GSE15471、GSE71989和GSE62165)验证了KIN17在胰腺癌中的上调,并利用公开的数据集(Gene Expression Profiling Interactive Analysis)确定了PI3K/Akt/mTOR通路与患者预后之间的关联。免疫组化检测KIN17与临床胰腺癌病理特征的关系。此外,研究表明,敲低KIN17可以抑制胰腺癌细胞的迁移和侵袭,并通过下调Vimentin和N - cadherin,上调E - cadherin,逆转胰腺癌细胞的上皮-间质转化。通过各种细胞实验,探索KIIN17在PI3K/AKT/mTOR活性中的作用。研究表明,抑制KIN17可通过PI3K/AKT/mTOR介导的自噬抑制胰腺癌细胞的迁移和侵袭。此外,与mTOR抑制联合,双重抑制可增强自噬,从而在胰腺癌中产生抗迁移和抗侵袭作用。综上所述,本研究提示KIN17可能具有致癌作用,并可作为胰腺癌的预后生物标志物。此外,KIN17可能被认为是一个潜在的治疗靶点,其敲除对胰腺癌具有抑制作用。
{"title":"Regulation of pancreatic cancer cells by suppressing KIN17 through the PI3K/AKT/mTOR signaling pathway.","authors":"Qiuyan Li, Yuxia Yang, Xiaocong Lin, Lok Ting Chu, Helian Chen, Linsong Chen, Jinjing Tang, Tao Zeng","doi":"10.3892/or.2025.8864","DOIUrl":"10.3892/or.2025.8864","url":null,"abstract":"<p><p>Pancreatic cancer is an aggressive tumor, which is often associated with a poor clinical prognosis and resistance to conventional chemotherapy. Therefore, there is a need to identify new therapeutic markers for pancreatic cancer. Although KIN17 is a highly expressed DNA‑ and RNA‑binding protein in a number of types of human cancer, its role in pancreatic cancer development, especially in relation to progression, is currently unknown. The present study verified the upregulation of KIN17 in pancreatic cancer using The Cancer Genome Atlas and Gene Expression Omnibus databases (GSE15471, GSE71989 and GSE62165), and identified an association between the PI3K/Akt/mTOR pathway and patient prognosis using publicly available datasets (Gene Expression Profiling Interactive Analysis). Immunohistochemistry was performed to determine the association between KIN17 and the pathological features of clinical pancreatic cancer samples. Furthermore, knockdown of KIN17 was shown to inhibit the migration and invasion of pancreatic cancer cells, and to reverse epithelial‑mesenchymal transition in pancreatic cancer cells through downregulation of Vimentin and N‑cadherin, and upregulation of E‑cadherin. Through various cellular experiments, the role of KIIN17 was explored in PI3K/AKT/mTOR activity. KIN17 inhibition was shown to suppress the migration and invasion of pancreatic cancer cells through PI3K/AKT/mTOR‑mediated autophagy. Furthermore, combined with mTOR inhibition, dual inhibition could enhance autophagy, leading to anti‑migratory and anti‑invasion effects in pancreatic cancer. In conclusion, the present study indicated that KIN17 may have a role in carcinogenesis and could serve as a prognostic biomarker of pancreatic cancer, owing to its high expression. In addition, KIN17 may be considered a potential therapeutic target with its knockdown having an inhibitory effect on pancreatic cancer.</p>","PeriodicalId":19527,"journal":{"name":"Oncology reports","volume":"53 2","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11736091/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142952637","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Regulation of the β‑catenin/LEF‑1 pathway by the siRNA knockdown of RUVBL1 expression inhibits breast cancer cell proliferation, migration and invasion. 通过 siRNA 敲除 RUVBL1 的表达来调节β-catenin/LEF-1 通路,从而抑制乳腺癌细胞的增殖、迁移和侵袭。
IF 3.8 3区 医学 Q2 ONCOLOGY Pub Date : 2025-02-01 Epub Date: 2024-12-13 DOI: 10.3892/or.2024.8855
Xin Zhang, Dingyuan Cui, Wei Sun, Guangfei Yang, Wen Wang, Chengrong Mi

RUVBL1 is a protein characterized by its DNA‑dependent ATPase activity and DNA deconjugating enzyme function. It is a member of the ATPase (AAA+) protein family associated with various cellular processes. Available research confirms that the expression of RUVBL1 is upregulated in breast cancer (BRCA) cell lines; however, the mechanisms underlying its functional role in BRCA remain unclear. The β‑catenin/lymphoid enhancer factor‑1 (LEF‑1) pathway plays a crucial role in the occurrence and development of BRCA. The aim of the present study was to investigate whether RUVBL1 regulates the proliferation, migration and invasion of BRCA cells by participating in the β‑catenin/LEF‑1 signaling pathway. Reverse transcription‑quantitative PCR (RT‑qPCR) and western blot analysis were employed to compare the RUVBL1 expression levels between normal mammary epithelial cells (MCF‑10a) and BRCA cell lines (MDA‑MB‑231 and MCF‑7). Scratch, Cell Counting Kit‑8 and Transwell assays were utilized to assess the effects of RUVBL1 knockdown on the proliferation, migration and invasion of BRCA cells. Following the downregulation of RUVBL1 expression in vitro, western blot analysis and RT‑qPCR were conducted to investigate its role in regulating the β‑catenin/LEF‑1 pathway. The aforementioned experiments proved that the knockdown of RUVBL1 expression inhibited BRCA cell proliferative, migratory and invasive capabilities, modulating the β‑catenin/LEF‑1 pathway. Collectively, the findings of the present study provide preliminarily confirmation that RUVBL1 participates in the molecular mechanisms of the β‑catenin signaling pathway, which may provide a novel target for BRCA treatment.

RUVBL1是一种具有DNA依赖性atp酶活性和DNA解偶联酶功能的蛋白质。它是atp酶(AAA+)蛋白家族的成员,与各种细胞过程相关。现有研究证实,RUVBL1在乳腺癌(BRCA)细胞系中表达上调;然而,其在BRCA中的功能作用机制尚不清楚。β - catenin/lymphoid enhancer factor - 1 (LEF - 1)通路在BRCA的发生发展中起着至关重要的作用。本研究旨在探讨RUVBL1是否通过参与β - catenin/LEF - 1信号通路调控BRCA细胞的增殖、迁移和侵袭。采用反转录定量PCR (RT - qPCR)和western blot分析比较正常乳腺上皮细胞(MCF - 10a)和BRCA细胞系(MDA - MB - 231和MCF - 7) RUVBL1的表达水平。使用Scratch、Cell Counting Kit‑8和Transwell检测来评估RUVBL1敲低对BRCA细胞增殖、迁移和侵袭的影响。在体外下调RUVBL1表达后,采用western blot和RT - qPCR方法研究其在β - catenin/LEF - 1通路中的调控作用。上述实验证明,RUVBL1表达下调可抑制BRCA细胞的增殖、迁移和侵袭能力,调节β - catenin/LEF - 1通路。综上所述,本研究结果初步证实RUVBL1参与β - catenin信号通路的分子机制,可能为BRCA的治疗提供新的靶点。
{"title":"Regulation of the β‑catenin/LEF‑1 pathway by the siRNA knockdown of RUVBL1 expression inhibits breast cancer cell proliferation, migration and invasion.","authors":"Xin Zhang, Dingyuan Cui, Wei Sun, Guangfei Yang, Wen Wang, Chengrong Mi","doi":"10.3892/or.2024.8855","DOIUrl":"10.3892/or.2024.8855","url":null,"abstract":"<p><p>RUVBL1 is a protein characterized by its DNA‑dependent ATPase activity and DNA deconjugating enzyme function. It is a member of the ATPase (AAA+) protein family associated with various cellular processes. Available research confirms that the expression of RUVBL1 is upregulated in breast cancer (BRCA) cell lines; however, the mechanisms underlying its functional role in BRCA remain unclear. The β‑catenin/lymphoid enhancer factor‑1 (LEF‑1) pathway plays a crucial role in the occurrence and development of BRCA. The aim of the present study was to investigate whether RUVBL1 regulates the proliferation, migration and invasion of BRCA cells by participating in the β‑catenin/LEF‑1 signaling pathway. Reverse transcription‑quantitative PCR (RT‑qPCR) and western blot analysis were employed to compare the RUVBL1 expression levels between normal mammary epithelial cells (MCF‑10a) and BRCA cell lines (MDA‑MB‑231 and MCF‑7). Scratch, Cell Counting Kit‑8 and Transwell assays were utilized to assess the effects of RUVBL1 knockdown on the proliferation, migration and invasion of BRCA cells. Following the downregulation of RUVBL1 expression <i>in vitro</i>, western blot analysis and RT‑qPCR were conducted to investigate its role in regulating the β‑catenin/LEF‑1 pathway. The aforementioned experiments proved that the knockdown of RUVBL1 expression inhibited BRCA cell proliferative, migratory and invasive capabilities, modulating the β‑catenin/LEF‑1 pathway. Collectively, the findings of the present study provide preliminarily confirmation that RUVBL1 participates in the molecular mechanisms of the β‑catenin signaling pathway, which may provide a novel target for BRCA treatment.</p>","PeriodicalId":19527,"journal":{"name":"Oncology reports","volume":"53 2","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11667213/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142818804","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Impact of miRNAs involved in the STAT3 signaling pathway on esophageal cancer (Review). STAT3信号通路中涉及的mirna对食管癌的影响(综述)。
IF 3.8 3区 医学 Q2 ONCOLOGY Pub Date : 2025-02-01 Epub Date: 2025-01-03 DOI: 10.3892/or.2024.8860
Ying Xiong, Yi-Fan Liu, Zhi-Hui Yang, Cong-Gai Huang

Esophageal cancer (ESCA) is a common tumor noted in the digestive tract, which is highly malignant due to unclear early symptoms and poor last‑stage treatment effects; its mortality rate is relatively high. MicroRNA (miR) and signal transducer and activator of transcription 3 (STAT3) are key components of cellular signaling pathways; their interaction forms a complex and intricate information network that controls several types of biological behaviors in the cells. In the tumor cell, these signal transduction pathways are abnormally active, indicating that the STAT3 signaling pathway mediated by miRs is involved in the progression of various cancer types. The present review introduces the biological characteristics of miR and STAT3 and their relationship with ESCA. It summarizes the regulation of ESCA by the miR and STAT3 signaling pathways and analyzes the effects of these pathways on proliferation, apoptosis, invasion, metastasis and immune escape of cancer cells, as well as the impact on patient survival and prognosis. The purpose of the present review is to assess the miR/STAT3 signaling pathway in ESCA, improve the understanding of the pathogenesis of ESCA and facilitate the identification of therapeutic targets for ESCA.

食管癌(ESCA)是一种常见的消化道肿瘤,早期症状不清,晚期治疗效果差,恶性程度高;它的死亡率相对较高。MicroRNA (miR)和信号转导和转录激活因子3 (STAT3)是细胞信号通路的关键组成部分;它们的相互作用形成了一个复杂而复杂的信息网络,控制着细胞中几种类型的生物行为。在肿瘤细胞中,这些信号转导通路异常活跃,表明由miRs介导的STAT3信号通路参与了多种癌症类型的进展。现就miR和STAT3的生物学特性及其与ESCA的关系作一综述。总结miR和STAT3信号通路对ESCA的调控,分析这些通路对癌细胞增殖、凋亡、侵袭、转移和免疫逃逸的影响,以及对患者生存和预后的影响。本文综述的目的是评估miR/STAT3信号通路在ESCA中的作用,提高对ESCA发病机制的认识,并有助于确定ESCA的治疗靶点。
{"title":"Impact of miRNAs involved in the STAT3 signaling pathway on esophageal cancer (Review).","authors":"Ying Xiong, Yi-Fan Liu, Zhi-Hui Yang, Cong-Gai Huang","doi":"10.3892/or.2024.8860","DOIUrl":"https://doi.org/10.3892/or.2024.8860","url":null,"abstract":"<p><p>Esophageal cancer (ESCA) is a common tumor noted in the digestive tract, which is highly malignant due to unclear early symptoms and poor last‑stage treatment effects; its mortality rate is relatively high. MicroRNA (miR) and signal transducer and activator of transcription 3 (STAT3) are key components of cellular signaling pathways; their interaction forms a complex and intricate information network that controls several types of biological behaviors in the cells. In the tumor cell, these signal transduction pathways are abnormally active, indicating that the STAT3 signaling pathway mediated by miRs is involved in the progression of various cancer types. The present review introduces the biological characteristics of miR and STAT3 and their relationship with ESCA. It summarizes the regulation of ESCA by the miR and STAT3 signaling pathways and analyzes the effects of these pathways on proliferation, apoptosis, invasion, metastasis and immune escape of cancer cells, as well as the impact on patient survival and prognosis. The purpose of the present review is to assess the miR/STAT3 signaling pathway in ESCA, improve the understanding of the pathogenesis of ESCA and facilitate the identification of therapeutic targets for ESCA.</p>","PeriodicalId":19527,"journal":{"name":"Oncology reports","volume":"53 2","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142922482","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Oncology reports
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:481959085
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