首页 > 最新文献

Cancer Cell International最新文献

英文 中文
Adipocytes promote metastasis of breast cancer by attenuating the FOXO1 effects and regulating copper homeostasis 脂肪细胞通过减弱 FOXO1 作用和调节铜平衡促进乳腺癌转移
IF 5.8 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-12 DOI: 10.1186/s12935-024-03433-y
Xiu Chen, Heda Zhang, Zheng Fang, Dandan Wang, Yuxin Song, Qian Zhang, Junchen Hou, Sujin Yang, Di Xu, Yinjiao Fei, Wei Zhang, Jian Zhang, Jinhai Tang, Lei Li
Obesity and the forkhead box O1(FOXO1) affect the survival of breast cancer patients, but the underlying mechanism remains unclear. We aimed to investigate the role of FOXO1 in obesity-associated-breast cancer. We screened 383 breast disease patients from the first affiliated hospital with Nanjing Medical University in 2020. We performed wound healing, transwell, matrigel assays to assess the metastatic ability of cancer cells. We adopted mRNAs sequencing to select the differentially expressed transcripts in breast cancer. We applied immunohistochemistry, western blot, tissue microarrays to assess the level of FOXO1 and epithelial-mesenchymal transition (EMT) pathways. We conducted bioinformatic analysis to investigate interactions between FOXO1 and miR-135b. We used fluorescence in situ hybridization, RT-qPCR to confirm the characteristics of circCNIH4. We conducted luciferase reporter assay, rescue experiments to investigate interactions between circCNIH4 and miR-135b. Obesity was positively correlated with the incidence and progression of breast cancer. Adipocytes enhanced the migration of breast cancer and attenuated the effects of FOXO1. MiR-135b was a binding gene of FOXO1 and was regulated by circCNIH4. CircCNIH4 exhibited antitumor activity in vitro and in vivo. Adipocytes might accelerate the progression of breast cancer by modulating FOXO1/miR-135b/ circCNIH4 /EMT axis and regulating copper homeostasis.
肥胖和叉头框O1(FOXO1)会影响乳腺癌患者的生存,但其潜在机制仍不清楚。我们旨在研究FOXO1在肥胖相关性乳腺癌中的作用。我们对南京医科大学第一附属医院2020年收治的383名乳腺疾病患者进行了筛查。我们通过伤口愈合、transwell和matrigel实验来评估癌细胞的转移能力。采用mRNA测序筛选乳腺癌中差异表达的转录本。我们采用免疫组化、Western 印迹和组织芯片来评估 FOXO1 和上皮-间质转化(EMT)通路的水平。我们进行了生物信息学分析,以研究 FOXO1 和 miR-135b 之间的相互作用。我们使用荧光原位杂交、RT-qPCR来确认circCNIH4的特征。我们进行了荧光素酶报告实验和拯救实验来研究 circCNIH4 和 miR-135b 之间的相互作用。肥胖与乳腺癌的发病和进展呈正相关。脂肪细胞增强了乳腺癌的迁移并减弱了FOXO1的作用。MiR-135b 是 FOXO1 的结合基因,并受 circCNIH4 的调控。CircCNIH4在体外和体内都具有抗肿瘤活性。脂肪细胞可能通过调节FOXO1/miR-135b/ circCNIH4/EMT轴和铜平衡加速乳腺癌的进展。
{"title":"Adipocytes promote metastasis of breast cancer by attenuating the FOXO1 effects and regulating copper homeostasis","authors":"Xiu Chen, Heda Zhang, Zheng Fang, Dandan Wang, Yuxin Song, Qian Zhang, Junchen Hou, Sujin Yang, Di Xu, Yinjiao Fei, Wei Zhang, Jian Zhang, Jinhai Tang, Lei Li","doi":"10.1186/s12935-024-03433-y","DOIUrl":"https://doi.org/10.1186/s12935-024-03433-y","url":null,"abstract":"Obesity and the forkhead box O1(FOXO1) affect the survival of breast cancer patients, but the underlying mechanism remains unclear. We aimed to investigate the role of FOXO1 in obesity-associated-breast cancer. We screened 383 breast disease patients from the first affiliated hospital with Nanjing Medical University in 2020. We performed wound healing, transwell, matrigel assays to assess the metastatic ability of cancer cells. We adopted mRNAs sequencing to select the differentially expressed transcripts in breast cancer. We applied immunohistochemistry, western blot, tissue microarrays to assess the level of FOXO1 and epithelial-mesenchymal transition (EMT) pathways. We conducted bioinformatic analysis to investigate interactions between FOXO1 and miR-135b. We used fluorescence in situ hybridization, RT-qPCR to confirm the characteristics of circCNIH4. We conducted luciferase reporter assay, rescue experiments to investigate interactions between circCNIH4 and miR-135b. Obesity was positively correlated with the incidence and progression of breast cancer. Adipocytes enhanced the migration of breast cancer and attenuated the effects of FOXO1. MiR-135b was a binding gene of FOXO1 and was regulated by circCNIH4. CircCNIH4 exhibited antitumor activity in vitro and in vivo. Adipocytes might accelerate the progression of breast cancer by modulating FOXO1/miR-135b/ circCNIH4 /EMT axis and regulating copper homeostasis.","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141946846","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Upregulation of MELK promotes chemoresistance and induces macrophage M2 polarization via CSF-1/JAK2/STAT3 pathway in gastric cancer. MELK的上调可促进胃癌的化疗耐药性,并通过CSF-1/JAK2/STAT3途径诱导巨噬细胞M2极化。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-12 DOI: 10.1186/s12935-024-03453-8
Pengfei Su, Tian Yu, Yingjing Zhang, Hongyun Huang, Moxi Chen, Can Cao, Weiming Kang, Yuqin Liu, Jianchun Yu

Background: Gastric cancer (GC) stands out as one of the most prevalent malignancies affecting the digestive system, characterized by a substantial incidence rate and mortality. Maternal embryonic leucine zipper kinase (MELK) has been implicated in the advancement of various cancer types and the modulation of the tumor microenvironment. This study aims to delve into the involvement of MELK in chemoresistance and the tumor microenvironment of GC.

Methods: The MELK expression was detected using quantitative real-time polymerase chain reaction (qRT-PCR), western blotting and immunohistochemistry. Lentiviral transfection was employed to establish stable cell lines with either overexpressed or silenced MELK. The impact of MELK on the chemoresistance of GC cells and the polarization of macrophages was investigated through in vitro and in vivo functional assays. Additionally, the correlation between MELK and the cytokines colony-stimulating factor 1 (CSF-1), as well as stromal macrophages, was analysed. The prognostic significance of MELK, CSF-1, and CD206 expression levels in clinical samples was further investigated.

Results: MELK was found to be highly expressed in chemoresistant GC cells and tissues. Furthermore, both in vitro and in vivo assays indicated that MELK overexpression conferred chemoresistance in GC cells. Additionally, MELK overexpression was observed to induce M2 macrophage polarization via the CSF-1/JAK2/STAT3 pathway, thereby contributing to chemoresistance within the tumor microenvironment. The expression of MELK in GC tissues from neoadjuvant chemotherapy patients correlated positively with CSF-1 and CD206. Moreover, patients with higher expression levels of MELK, CSF-1, or CD206 exhibited significantly shorter OS and DFS rates.

Conclusions: Our investigation underscores the critical role of MELK in promoting chemoresistance and inducing M2 macrophage polarization in GC. It proposes novel targets and methods for the treatment of GC, as well as prognostic factors for neoadjuvant chemotherapy.

背景:胃癌(GC)是影响消化系统的最常见恶性肿瘤之一,发病率和死亡率都很高。母体胚胎亮氨酸拉链激酶(MELK)与各种癌症类型的发展和肿瘤微环境的调节有关。本研究旨在探讨MELK参与GC化疗耐药性和肿瘤微环境的情况:方法:采用实时定量聚合酶链反应(qRT-PCR)、免疫印迹法和免疫组化法检测MELK的表达。采用慢病毒转染技术建立过表达或沉默MELK的稳定细胞系。通过体外和体内功能测试,研究了MELK对GC细胞化疗抗性和巨噬细胞极化的影响。此外,还分析了MELK与细胞因子集落刺激因子1(CSF-1)以及基质巨噬细胞之间的相关性。还进一步研究了临床样本中MELK、CSF-1和CD206表达水平的预后意义:结果:研究发现,MELK在化疗耐药的GC细胞和组织中高度表达。此外,体外和体内试验均表明,MELK 的过度表达会使 GC 细胞产生化疗耐药性。此外,还观察到MELK过表达可通过CSF-1/JAK2/STAT3途径诱导M2巨噬细胞极化,从而导致肿瘤微环境中的化疗抵抗。新辅助化疗患者GC组织中MELK的表达与CSF-1和CD206呈正相关。此外,MELK、CSF-1或CD206表达水平较高的患者的OS和DFS率明显较短:我们的研究强调了 MELK 在促进 GC 化疗耐药性和诱导 M2 巨噬细胞极化中的关键作用。结论:我们的研究强调了 MELK 在促进 GC 化疗耐药和诱导 M2 巨噬细胞极化中的关键作用,并提出了治疗 GC 的新靶点和方法,以及新辅助化疗的预后因素。
{"title":"Upregulation of MELK promotes chemoresistance and induces macrophage M2 polarization via CSF-1/JAK2/STAT3 pathway in gastric cancer.","authors":"Pengfei Su, Tian Yu, Yingjing Zhang, Hongyun Huang, Moxi Chen, Can Cao, Weiming Kang, Yuqin Liu, Jianchun Yu","doi":"10.1186/s12935-024-03453-8","DOIUrl":"10.1186/s12935-024-03453-8","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer (GC) stands out as one of the most prevalent malignancies affecting the digestive system, characterized by a substantial incidence rate and mortality. Maternal embryonic leucine zipper kinase (MELK) has been implicated in the advancement of various cancer types and the modulation of the tumor microenvironment. This study aims to delve into the involvement of MELK in chemoresistance and the tumor microenvironment of GC.</p><p><strong>Methods: </strong>The MELK expression was detected using quantitative real-time polymerase chain reaction (qRT-PCR), western blotting and immunohistochemistry. Lentiviral transfection was employed to establish stable cell lines with either overexpressed or silenced MELK. The impact of MELK on the chemoresistance of GC cells and the polarization of macrophages was investigated through in vitro and in vivo functional assays. Additionally, the correlation between MELK and the cytokines colony-stimulating factor 1 (CSF-1), as well as stromal macrophages, was analysed. The prognostic significance of MELK, CSF-1, and CD206 expression levels in clinical samples was further investigated.</p><p><strong>Results: </strong>MELK was found to be highly expressed in chemoresistant GC cells and tissues. Furthermore, both in vitro and in vivo assays indicated that MELK overexpression conferred chemoresistance in GC cells. Additionally, MELK overexpression was observed to induce M2 macrophage polarization via the CSF-1/JAK2/STAT3 pathway, thereby contributing to chemoresistance within the tumor microenvironment. The expression of MELK in GC tissues from neoadjuvant chemotherapy patients correlated positively with CSF-1 and CD206. Moreover, patients with higher expression levels of MELK, CSF-1, or CD206 exhibited significantly shorter OS and DFS rates.</p><p><strong>Conclusions: </strong>Our investigation underscores the critical role of MELK in promoting chemoresistance and inducing M2 macrophage polarization in GC. It proposes novel targets and methods for the treatment of GC, as well as prognostic factors for neoadjuvant chemotherapy.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11320770/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141970671","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
TTI-101 targets STAT3/c-Myc signaling pathway to suppress cervical cancer progression: an integrated experimental and computational analysis. TTI-101靶向STAT3/c-Myc信号通路抑制宫颈癌进展:实验与计算综合分析
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-12 DOI: 10.1186/s12935-024-03463-6
Yi Li, Yuyan Dong

Background: Cervical cancer (CC) is a significant global health concern, demanding the consideration of novel therapeutic strategies. The signal transducer and activator of transcription 3 (STAT3) pathway has been implicated in cancer progression and is a potential target for therapeutic intervention. This study aimed to explore the therapeutic potential of TTI-101, a small molecule STAT3 inhibitor, in CC and investigate its underlying mechanisms.

Methods: Molecular docking studies and molecular dynamics simulations were performed to explore the binding interaction between TTI-101 and STAT3 and assess the stability of the STAT3-TTI-101 complex. Cell viability assays, wound healing assays, colony formation assays, flow cytometry analysis, and gene expression analysis were conducted. In vivo xenograft models were used to assess the antitumor efficacy of TTI-101.

Results: The in silico analysis shows a stable binding interaction between TTI-101 and STAT3. TTI-101 treatment inhibits cell viability, clonogenic ability, and cell migration in CC cells. Furthermore, TTI-101 induces apoptosis and cell cycle arrest. Analysis of apoptosis-related markers demonstrated dysregulation of Bax, Bcl-2, and Caspase-3 upon TTI-101 treatment. Moreover, TTI-101 caused G2/M phase arrest accompanied by a decrease in CDK1 and Cyclin B1 at mRNA levels. In the xenograft model, TTI-101 significantly inhibited tumor growth without adverse effects on body weight.

Conclusion: TTI-101 exhibited anticancer effects by targeting the STAT3/c-Myc signaling pathway, inducing cell cycle arrest, and promoting apoptosis in CC cells. These findings provide valuable insights into the development of novel therapeutic strategies for cervical cancer. Further investigation is warranted to validate the clinical application of TTI-101.

背景:宫颈癌(CC)是全球关注的重大健康问题,需要考虑新的治疗策略。信号转导和转录激活因子 3(STAT3)通路与癌症进展有牵连,是治疗干预的潜在靶点。本研究旨在探索小分子 STAT3 抑制剂 TTI-101 在 CC 中的治疗潜力,并研究其潜在机制:方法:通过分子对接研究和分子动力学模拟探索 TTI-101 与 STAT3 之间的结合相互作用,并评估 STAT3-TTI-101 复合物的稳定性。还进行了细胞活力测定、伤口愈合测定、集落形成测定、流式细胞仪分析和基因表达分析。体内异种移植模型用于评估 TTI-101 的抗肿瘤疗效:硅学分析表明,TTI-101与STAT3之间存在稳定的结合相互作用。TTI-101可抑制CC细胞的活力、克隆生成能力和细胞迁移。此外,TTI-101 还能诱导细胞凋亡和细胞周期停滞。对细胞凋亡相关标志物的分析表明,TTI-101 处理后,Bax、Bcl-2 和 Caspase-3 出现失调。此外,TTI-101 还会导致 G2/M 期停滞,并伴随着 CDK1 和 Cyclin B1 mRNA 水平的下降。在异种移植模型中,TTI-101能显著抑制肿瘤生长,且对体重无不良影响:结论:TTI-101通过靶向STAT3/c-Myc信号通路、诱导细胞周期停滞和促进CC细胞凋亡而发挥抗癌作用。这些发现为宫颈癌新型治疗策略的开发提供了宝贵的见解。要验证TTI-101的临床应用,还需要进一步的研究。
{"title":"TTI-101 targets STAT3/c-Myc signaling pathway to suppress cervical cancer progression: an integrated experimental and computational analysis.","authors":"Yi Li, Yuyan Dong","doi":"10.1186/s12935-024-03463-6","DOIUrl":"10.1186/s12935-024-03463-6","url":null,"abstract":"<p><strong>Background: </strong>Cervical cancer (CC) is a significant global health concern, demanding the consideration of novel therapeutic strategies. The signal transducer and activator of transcription 3 (STAT3) pathway has been implicated in cancer progression and is a potential target for therapeutic intervention. This study aimed to explore the therapeutic potential of TTI-101, a small molecule STAT3 inhibitor, in CC and investigate its underlying mechanisms.</p><p><strong>Methods: </strong>Molecular docking studies and molecular dynamics simulations were performed to explore the binding interaction between TTI-101 and STAT3 and assess the stability of the STAT3-TTI-101 complex. Cell viability assays, wound healing assays, colony formation assays, flow cytometry analysis, and gene expression analysis were conducted. In vivo xenograft models were used to assess the antitumor efficacy of TTI-101.</p><p><strong>Results: </strong>The in silico analysis shows a stable binding interaction between TTI-101 and STAT3. TTI-101 treatment inhibits cell viability, clonogenic ability, and cell migration in CC cells. Furthermore, TTI-101 induces apoptosis and cell cycle arrest. Analysis of apoptosis-related markers demonstrated dysregulation of Bax, Bcl-2, and Caspase-3 upon TTI-101 treatment. Moreover, TTI-101 caused G2/M phase arrest accompanied by a decrease in CDK1 and Cyclin B1 at mRNA levels. In the xenograft model, TTI-101 significantly inhibited tumor growth without adverse effects on body weight.</p><p><strong>Conclusion: </strong>TTI-101 exhibited anticancer effects by targeting the STAT3/c-Myc signaling pathway, inducing cell cycle arrest, and promoting apoptosis in CC cells. These findings provide valuable insights into the development of novel therapeutic strategies for cervical cancer. Further investigation is warranted to validate the clinical application of TTI-101.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11320917/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141970670","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Enhancing mutation detection in multiple myeloma with an error-corrected ultra-sensitive NGS assay without plasma cell enrichment 无需浆细胞富集的误差校正超灵敏 NGS 检测法可提高多发性骨髓瘤的突变检测能力
IF 5.8 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-12 DOI: 10.1186/s12935-024-03470-7
Jin Ju Kim, Soo-Jeong Kim, Seoyoung Lim, Seung-Tae Lee, Jong Rak Choi, Saeam Shin, Doh Yu Hwang
Risk stratification in multiple myeloma (MM) patients is crucial, and molecular genetic studies play a significant role in achieving this objective. Enrichment of plasma cells for next-generation sequencing (NGS) analysis has been employed to enhance detection sensitivity. However, these methods often come with limitations, such as high costs and low throughput. In this study, we explore the use of an error-corrected ultrasensitive NGS assay called positional indexing sequencing (PiSeq-MM). This assay can detect somatic mutations in MM patients without relying on plasma cell enrichment. Diagnostic bone marrow aspirates (BMAs) and blood samples from 14 MM patients were used for exploratory and validation sets. PiSeq-MM successfully detected somatic mutations in all BMAs, outperforming conventional NGS using plasma cells. It also identified 38 low-frequency mutations that were missed by conventional NGS, enhancing detection sensitivity below the 5% analytical threshold. When tested in an actual clinical environment, plasma cell enrichment failed in most BMAs (14/16), but the PiSeq-MM enabled mutation detection in all BMAs. There was concordance between PiSeq-MM using BMAs and ctDNA analysis in paired blood samples. This research provides valuable insights into the genetic landscape of MM and highlights the advantages of error-corrected NGS for detecting low-frequency mutations. Although the current standard method for mutation analysis is plasma cell-enriched BMAs, total BMA or ctDNA testing with error correction is a viable alternative when plasma cell enrichment is not feasible.
对多发性骨髓瘤(MM)患者进行风险分层至关重要,而分子遗传学研究在实现这一目标方面发挥着重要作用。为了提高检测灵敏度,人们采用了富集浆细胞进行下一代测序(NGS)分析的方法。然而,这些方法往往存在成本高、通量低等局限性。在本研究中,我们探索了一种名为位置索引测序(PiSeq-MM)的误差校正超灵敏 NGS 检测方法。这种检测方法可以检测 MM 患者的体细胞突变,而无需依赖浆细胞富集。14 名 MM 患者的诊断性骨髓抽吸物(BMA)和血液样本被用于探索集和验证集。PiSeq-MM 成功检测到了所有 BMA 中的体细胞突变,优于使用浆细胞的传统 NGS。它还发现了 38 个被传统 NGS 遗漏的低频突变,提高了低于 5% 分析阈值的检测灵敏度。在实际临床环境中进行测试时,大多数 BMA(14/16)的血浆细胞富集失败,但 PiSeq-MM 能够在所有 BMA 中检测到突变。使用 BMA 的 PiSeq-MM 与配对血液样本中的 ctDNA 分析结果一致。这项研究为了解 MM 的遗传情况提供了有价值的见解,并凸显了误差校正 NGS 在检测低频突变方面的优势。虽然目前突变分析的标准方法是血浆细胞富集的 BMA,但当血浆细胞富集不可行时,进行误差校正的总 BMA 或 ctDNA 检测是一种可行的替代方法。
{"title":"Enhancing mutation detection in multiple myeloma with an error-corrected ultra-sensitive NGS assay without plasma cell enrichment","authors":"Jin Ju Kim, Soo-Jeong Kim, Seoyoung Lim, Seung-Tae Lee, Jong Rak Choi, Saeam Shin, Doh Yu Hwang","doi":"10.1186/s12935-024-03470-7","DOIUrl":"https://doi.org/10.1186/s12935-024-03470-7","url":null,"abstract":"Risk stratification in multiple myeloma (MM) patients is crucial, and molecular genetic studies play a significant role in achieving this objective. Enrichment of plasma cells for next-generation sequencing (NGS) analysis has been employed to enhance detection sensitivity. However, these methods often come with limitations, such as high costs and low throughput. In this study, we explore the use of an error-corrected ultrasensitive NGS assay called positional indexing sequencing (PiSeq-MM). This assay can detect somatic mutations in MM patients without relying on plasma cell enrichment. Diagnostic bone marrow aspirates (BMAs) and blood samples from 14 MM patients were used for exploratory and validation sets. PiSeq-MM successfully detected somatic mutations in all BMAs, outperforming conventional NGS using plasma cells. It also identified 38 low-frequency mutations that were missed by conventional NGS, enhancing detection sensitivity below the 5% analytical threshold. When tested in an actual clinical environment, plasma cell enrichment failed in most BMAs (14/16), but the PiSeq-MM enabled mutation detection in all BMAs. There was concordance between PiSeq-MM using BMAs and ctDNA analysis in paired blood samples. This research provides valuable insights into the genetic landscape of MM and highlights the advantages of error-corrected NGS for detecting low-frequency mutations. Although the current standard method for mutation analysis is plasma cell-enriched BMAs, total BMA or ctDNA testing with error correction is a viable alternative when plasma cell enrichment is not feasible.","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141969562","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Synergistic antitumor effect of liposomal-based formulations of olaparib and topotecan in primary epithelial ovarian cancer cells. 奥拉帕利和托泊替康脂质体制剂在原发性上皮卵巢癌细胞中的协同抗肿瘤作用
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-12 DOI: 10.1186/s12935-024-03469-0
Aleksandra Romaniuk-Drapala, Paulina Skupin-Mrugalska, Olga Garbuzenko, Arash Hatefi, Tamara Minko

Background: Olaparib is a PARP inhibitor inducing synthetic lethality in tumors with deficient homologous recombination (HRD) caused by BRCA1/2 mutations. The FDA has approved monotherapy for first-line platinum-sensitive, recurrent high-grade epithelial ovarian cancer. Combination therapy alongside DNA-damaging therapeutics is a promising solution to overcome the limited efficacy in patients with HRD. The present study was designed to develop topotecan- and olaparib-loaded liposomes (TLL and OLL) and assess the effectiveness of their combination in patient-derived ovarian cancer samples.

Methods: We used HEOC, four clear-cell tumors (EOC 1-4), malignant ascites, and an OCI-E1p endometrioid primary ovarian cancer cell line and performed NGS analysis of BRCA1/2 mutation status. Antiproliferative activity was determined with the MTT assay. The Chou-Talalay algorithm was used to investigate the in vitro pharmacodynamic interactions of TLLs and OLLs.

Results: The OLL showed significantly higher efficacy in all ovarian cancer types with wild-type BRCA1/2 than a conventional formulation, suggesting potential for increased in vivo efficacy. The TLL revealed substantially higher toxicity to EOC 1, EOC 3, ascites and lower toxicity to HEOC than the standard formulation, suggesting better therapeutic efficacy and safety profile. The combination of studied compounds showed a higher reduction in cell viability than drugs used individually, demonstrating a synergistic antitumor effect at most of the selected concentrations.

Conclusions: The concentration-dependent response of different ovarian cancer cell types to combination therapy confirms the need for in vitro optimization to maximize drug cytotoxicity. The OLL and TLL combination is a promising formulation for further animal studies, especially for eliminating epithelial ovarian cancer with wild-type BRCA1/2.

背景:奥拉帕利(Olaparib)是一种PARP抑制剂,可诱导因BRCA1/2基因突变导致同源重组(HRD)缺陷的肿瘤合成致死。美国食品和药物管理局(FDA)已批准将单药疗法用于对铂敏感的复发性高级别上皮性卵巢癌的一线治疗。DNA损伤疗法与联合疗法是克服HRD患者有限疗效的一种有希望的解决方案。本研究旨在开发托泊替康和奥拉帕利脂质体(TLL 和 OLL),并评估它们在卵巢癌患者样本中的联合治疗效果:我们使用了 HEOC、四种透明细胞瘤(EOC 1-4)、恶性腹水和一种 OCI-E1p 子宫内膜原发性卵巢癌细胞系,并对 BRCA1/2 基因突变状态进行了 NGS 分析。抗增殖活性采用 MTT 法测定。采用 Chou-Talalay 算法研究了 TLL 和 OLL 的体外药效学相互作用:结果:OLL对所有野生型BRCA1/2卵巢癌类型的疗效均明显高于传统制剂,这表明OLL具有提高体内疗效的潜力。与标准制剂相比,TLL 对 EOC 1、EOC 3 和腹水的毒性要高得多,而对 HEOC 的毒性要低得多,这表明它具有更好的疗效和安全性。与单独使用的药物相比,所研究化合物的复方制剂能更有效地降低细胞活力,这表明在大多数选定浓度下,复方制剂具有协同抗肿瘤作用:结论:不同卵巢癌细胞类型对联合疗法的反应与浓度有关,这证明有必要进行体外优化,以最大限度地提高药物的细胞毒性。OLL和TLL联合疗法是一种很有前景的制剂,可用于进一步的动物实验,尤其是消除野生型BRCA1/2上皮性卵巢癌。
{"title":"Synergistic antitumor effect of liposomal-based formulations of olaparib and topotecan in primary epithelial ovarian cancer cells.","authors":"Aleksandra Romaniuk-Drapala, Paulina Skupin-Mrugalska, Olga Garbuzenko, Arash Hatefi, Tamara Minko","doi":"10.1186/s12935-024-03469-0","DOIUrl":"10.1186/s12935-024-03469-0","url":null,"abstract":"<p><strong>Background: </strong>Olaparib is a PARP inhibitor inducing synthetic lethality in tumors with deficient homologous recombination (HRD) caused by BRCA1/2 mutations. The FDA has approved monotherapy for first-line platinum-sensitive, recurrent high-grade epithelial ovarian cancer. Combination therapy alongside DNA-damaging therapeutics is a promising solution to overcome the limited efficacy in patients with HRD. The present study was designed to develop topotecan- and olaparib-loaded liposomes (TLL and OLL) and assess the effectiveness of their combination in patient-derived ovarian cancer samples.</p><p><strong>Methods: </strong>We used HEOC, four clear-cell tumors (EOC 1-4), malignant ascites, and an OCI-E1p endometrioid primary ovarian cancer cell line and performed NGS analysis of BRCA1/2 mutation status. Antiproliferative activity was determined with the MTT assay. The Chou-Talalay algorithm was used to investigate the in vitro pharmacodynamic interactions of TLLs and OLLs.</p><p><strong>Results: </strong>The OLL showed significantly higher efficacy in all ovarian cancer types with wild-type BRCA1/2 than a conventional formulation, suggesting potential for increased in vivo efficacy. The TLL revealed substantially higher toxicity to EOC 1, EOC 3, ascites and lower toxicity to HEOC than the standard formulation, suggesting better therapeutic efficacy and safety profile. The combination of studied compounds showed a higher reduction in cell viability than drugs used individually, demonstrating a synergistic antitumor effect at most of the selected concentrations.</p><p><strong>Conclusions: </strong>The concentration-dependent response of different ovarian cancer cell types to combination therapy confirms the need for in vitro optimization to maximize drug cytotoxicity. The OLL and TLL combination is a promising formulation for further animal studies, especially for eliminating epithelial ovarian cancer with wild-type BRCA1/2.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11320834/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141970669","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Retraction Note: Circular RNA hsa_circ_0000517 modulates hepatocellular carcinoma advancement via the miR-326/SMAD6 axis. 撤稿说明:环状 RNA hsa_circ_0000517 通过 miR-326/SMAD6 轴调节肝细胞癌的发展。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-09 DOI: 10.1186/s12935-024-03474-3
Shuwei He, Zhengwu Guo, Qian Kang, Xu Wang, Xingmin Han
{"title":"Retraction Note: Circular RNA hsa_circ_0000517 modulates hepatocellular carcinoma advancement via the miR-326/SMAD6 axis.","authors":"Shuwei He, Zhengwu Guo, Qian Kang, Xu Wang, Xingmin Han","doi":"10.1186/s12935-024-03474-3","DOIUrl":"10.1186/s12935-024-03474-3","url":null,"abstract":"","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11316384/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141911854","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Identification of POU4F1 as a novel prognostic biomarker and therapeutic target in esophageal squamous cell carcinoma. 将 POU4F1 鉴定为食管鳞状细胞癌的新型预后生物标记物和治疗靶点。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-09 DOI: 10.1186/s12935-024-03471-6
Nan Li, Siying Chen, Xiao Wang, Boqing Zhang, Boning Zeng, Chao Sun, Kai Zheng, Qiuling Chen, Shaoxiang Wang

Background: Esophageal cancer is a significant global health concern, ranking seventh in incidence and sixth in mortality. It encompasses two pathological types: esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma, with ESCC being more prevalent globally and associated with higher mortality rates. The POU (Pit-Oct-Unc) domain family transcription factors, comprising 15 members, play important roles in embryonic development and organ formation. Aberrant expression of POUs has been observed in several human cancers, influencing cell proliferation, tumor invasion, and drug resistance. However, their specific role in ESCC remains unknown.

Methods: We analyzed TCGA and GEO databases to assess POUs expression in ESCC tissues. Kaplan-Meier and ROC analyses were used to evaluate the prognostic value of POUs. Gene Set Enrichment Analysis and Protein-Protein interaction network were used to explore the potential pathway. Functional assays (Cell Counting Kit-8, EdU Staining assay, and cloning formation assay) and mechanism analyses (RNA-seq, flow cytometry, and Western blot) were conducted to determine the effects of POU4F1 knockdown on ESCC cell phenotypes and signaling pathways.

Results: POU4F1 and POU6F2 were upregulated in various cancer tissues, including ESCC, compared to normal tissues. POU4F1 expression was significantly correlated with patient survival and superior to previous models (AUC = 0.776). Knockdown of POU4F1 inhibited ESCC cell proliferation and affected cell cycle, autophagy, and DNA damage pathways in ESCC cells.

Conclusion: POU4F1 is a novel and promising prognostic and therapeutic target for ESCC patients, providing insights into potential treatment strategies.

背景:食管癌是全球关注的重大健康问题,发病率排名第七,死亡率排名第六。它包括两种病理类型:食管鳞状细胞癌(ESCC)和食管腺癌,其中食管鳞状细胞癌在全球发病率更高,死亡率也更高。POU(Pit-Oct-Unc)结构域家族转录因子共有 15 个成员,在胚胎发育和器官形成过程中发挥着重要作用。在几种人类癌症中都观察到了 POU 的异常表达,影响着细胞增殖、肿瘤侵袭和耐药性。然而,它们在 ESCC 中的具体作用仍然未知:我们分析了TCGA和GEO数据库,以评估POUs在ESCC组织中的表达。方法:我们分析了TCGA和GEO数据库,评估了POUs在ESCC组织中的表达,并使用Kaplan-Meier和ROC分析评估了POUs的预后价值。基因组富集分析(Gene Set Enrichment Analysis)和蛋白-蛋白相互作用网络(Protein-Protein interaction network)用于探索潜在的通路。通过功能检测(细胞计数试剂盒-8、EdU染色检测和克隆形成检测)和机制分析(RNA-seq、流式细胞术和Western印迹)确定POU4F1敲除对ESCC细胞表型和信号通路的影响:结果:与正常组织相比,POU4F1和POU6F2在包括ESCC在内的各种癌症组织中上调。POU4F1的表达与患者的生存期有明显相关性,且优于之前的模型(AUC = 0.776)。POU4F1的敲除抑制了ESCC细胞的增殖,并影响了ESCC细胞的细胞周期、自噬和DNA损伤途径:结论:POU4F1是ESCC患者的一个新的、有希望的预后和治疗靶点,为潜在的治疗策略提供了启示。
{"title":"Identification of POU4F1 as a novel prognostic biomarker and therapeutic target in esophageal squamous cell carcinoma.","authors":"Nan Li, Siying Chen, Xiao Wang, Boqing Zhang, Boning Zeng, Chao Sun, Kai Zheng, Qiuling Chen, Shaoxiang Wang","doi":"10.1186/s12935-024-03471-6","DOIUrl":"10.1186/s12935-024-03471-6","url":null,"abstract":"<p><strong>Background: </strong>Esophageal cancer is a significant global health concern, ranking seventh in incidence and sixth in mortality. It encompasses two pathological types: esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma, with ESCC being more prevalent globally and associated with higher mortality rates. The POU (Pit-Oct-Unc) domain family transcription factors, comprising 15 members, play important roles in embryonic development and organ formation. Aberrant expression of POUs has been observed in several human cancers, influencing cell proliferation, tumor invasion, and drug resistance. However, their specific role in ESCC remains unknown.</p><p><strong>Methods: </strong>We analyzed TCGA and GEO databases to assess POUs expression in ESCC tissues. Kaplan-Meier and ROC analyses were used to evaluate the prognostic value of POUs. Gene Set Enrichment Analysis and Protein-Protein interaction network were used to explore the potential pathway. Functional assays (Cell Counting Kit-8, EdU Staining assay, and cloning formation assay) and mechanism analyses (RNA-seq, flow cytometry, and Western blot) were conducted to determine the effects of POU4F1 knockdown on ESCC cell phenotypes and signaling pathways.</p><p><strong>Results: </strong>POU4F1 and POU6F2 were upregulated in various cancer tissues, including ESCC, compared to normal tissues. POU4F1 expression was significantly correlated with patient survival and superior to previous models (AUC = 0.776). Knockdown of POU4F1 inhibited ESCC cell proliferation and affected cell cycle, autophagy, and DNA damage pathways in ESCC cells.</p><p><strong>Conclusion: </strong>POU4F1 is a novel and promising prognostic and therapeutic target for ESCC patients, providing insights into potential treatment strategies.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11316379/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141911853","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Comprehensive analysis, diagnosis, prognosis, and cordycepin (CD) regulations for GSDME expressions in pan-cancers. 泛癌中 GSDME 表达的综合分析、诊断、预后和虫草素 (CD) 调节。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-08 DOI: 10.1186/s12935-024-03467-2
Jiewen Fu, Dabing Li, Lianmei Zhang, Mazaher Maghsoudloo, Jingliang Cheng, Junjiang Fu

The Gasdermin E gene (GSDME) plays roles in deafness and cancers. However, the roles and mechanisms in cancers are complex, and the same gene exhibits different mechanisms and actions in different types of cancers. Online databases, such as GEPIA2, cBioPortal, and DNMIVD, were used to comprehensively analyze GSDME profiles, DNA methylations, mutations, diagnosis, and prognosis in patients with tumor tissues and matched healthy tissues. Western blotting and RT-PCR were used to monitor the regulation of GSDME by Cordycepin (CD) in cancer cell lines. We revealed that GSDME expression is significantly upregulated in eight cancers (ACC, DLBC, GBM, HNSC, LGG, PAAD, SKCM, and THYM) and significantly downregulated in seven cancers (COAD, KICH, LAML, OV, READ, UCES, and UCS). The overall survival was longer only in ACC, but shorter in four cancers, including COAD, KIRC, LIHC, and STAD, when GSDME was highly expressed in cancers compared with the corresponding normal tissues. Moreover, the high expression of GSDME was negatively correlated with the poor prognosis of ACC, while the low expression of GSDME was negatively correlated with the poor prognosis of COAD, suggesting that GSDME might serve as a good prognostic factor in these two cancer types. Accordingly, results indicated that the DNA methylations of those 7 CpG sites constitute a potentially effective signature to distinguish different tumors from adjacent healthy tissues. Gene mutations for GSDME were frequently observed in a variety of tumors, with UCES having the highest frequency. Moreover, CD treatment inhibited GSDME expression in different cancer cell lines, while overexpression of GSDME promoted cell migration and invasion. Thus, we have systematically and successfully clarified the GSDME expression profiles, diagnostic values, and prognostic values in pan-cancers. Targeting GSDME with CD implies therapeutic significance and a mechanism for antitumor roles in some types of cancers via increasing the sensitivity of chemotherapy. Altogether, our study may provide a strategy and biomarker for clinical diagnosis, prognostics, and treatment of cancers by targeting GSDME.

Gasdermin E 基因(GSDME)在耳聋和癌症中发挥作用。然而,该基因在癌症中的作用和机制十分复杂,同一基因在不同类型的癌症中表现出不同的机制和作用。研究人员利用GEPIA2、cBioPortal和DNMIVD等在线数据库,全面分析了肿瘤患者组织和匹配健康组织的GSDME图谱、DNA甲基化、突变、诊断和预后。我们还利用 Western 印迹和 RT-PCR 技术监测了虫草素(CD)对肿瘤细胞株中 GSDME 的调控。我们发现,GSDME在8种癌症(ACC、DLBC、GBM、HNSC、LGG、PAAD、SKCM和THYM)中表达明显上调,在7种癌症(COAD、KICH、LAML、OV、READ、UCES和UCS)中表达明显下调。与相应的正常组织相比,当 GSDME 在癌症中高表达时,只有 ACC 的总生存期较长,而 COAD、KIRC、LIHC 和 STAD 等四种癌症的总生存期较短。此外,GSDME的高表达与ACC的不良预后呈负相关,而GSDME的低表达与COAD的不良预后呈负相关,这表明GSDME可能是这两种癌症的良好预后因素。因此,研究结果表明,这7个CpG位点的DNA甲基化可能是区分不同肿瘤和邻近健康组织的有效特征。GSDME基因突变在多种肿瘤中均有发现,其中UCES的频率最高。此外,CD治疗抑制了GSDME在不同癌细胞系中的表达,而GSDME的过表达则促进了细胞的迁移和侵袭。因此,我们成功地系统阐明了泛癌中GSDME的表达谱、诊断价值和预后价值。用CD靶向GSDME意味着治疗意义,以及通过提高化疗敏感性在某些类型癌症中发挥抗肿瘤作用的机制。总之,我们的研究可能会通过靶向GSDME为癌症的临床诊断、预后和治疗提供一种策略和生物标志物。
{"title":"Comprehensive analysis, diagnosis, prognosis, and cordycepin (CD) regulations for GSDME expressions in pan-cancers.","authors":"Jiewen Fu, Dabing Li, Lianmei Zhang, Mazaher Maghsoudloo, Jingliang Cheng, Junjiang Fu","doi":"10.1186/s12935-024-03467-2","DOIUrl":"10.1186/s12935-024-03467-2","url":null,"abstract":"<p><p>The Gasdermin E gene (GSDME) plays roles in deafness and cancers. However, the roles and mechanisms in cancers are complex, and the same gene exhibits different mechanisms and actions in different types of cancers. Online databases, such as GEPIA2, cBioPortal, and DNMIVD, were used to comprehensively analyze GSDME profiles, DNA methylations, mutations, diagnosis, and prognosis in patients with tumor tissues and matched healthy tissues. Western blotting and RT-PCR were used to monitor the regulation of GSDME by Cordycepin (CD) in cancer cell lines. We revealed that GSDME expression is significantly upregulated in eight cancers (ACC, DLBC, GBM, HNSC, LGG, PAAD, SKCM, and THYM) and significantly downregulated in seven cancers (COAD, KICH, LAML, OV, READ, UCES, and UCS). The overall survival was longer only in ACC, but shorter in four cancers, including COAD, KIRC, LIHC, and STAD, when GSDME was highly expressed in cancers compared with the corresponding normal tissues. Moreover, the high expression of GSDME was negatively correlated with the poor prognosis of ACC, while the low expression of GSDME was negatively correlated with the poor prognosis of COAD, suggesting that GSDME might serve as a good prognostic factor in these two cancer types. Accordingly, results indicated that the DNA methylations of those 7 CpG sites constitute a potentially effective signature to distinguish different tumors from adjacent healthy tissues. Gene mutations for GSDME were frequently observed in a variety of tumors, with UCES having the highest frequency. Moreover, CD treatment inhibited GSDME expression in different cancer cell lines, while overexpression of GSDME promoted cell migration and invasion. Thus, we have systematically and successfully clarified the GSDME expression profiles, diagnostic values, and prognostic values in pan-cancers. Targeting GSDME with CD implies therapeutic significance and a mechanism for antitumor roles in some types of cancers via increasing the sensitivity of chemotherapy. Altogether, our study may provide a strategy and biomarker for clinical diagnosis, prognostics, and treatment of cancers by targeting GSDME.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11312966/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141905929","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Harnessing exosomes as cancer biomarkers in clinical oncology. 在临床肿瘤学中利用外泌体作为癌症生物标记物。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-07 DOI: 10.1186/s12935-024-03464-5
Subhrojyoti Ghosh, Ramya Lakshmi Rajendran, Atharva A Mahajan, Ankita Chowdhury, Aishi Bera, Sudeepta Guha, Kashmira Chakraborty, Rajanyaa Chowdhury, Aritra Paul, Shreya Jha, Anuvab Dey, Amit Dubey, Sukhamoy Gorai, Purbasha Das, Chae Moon Hong, Anand Krishnan, Prakash Gangadaran, Byeong-Cheol Ahn

Exosomes are extracellular vesicles well known for facilitating cell-to-cell communication by distributing essential macromolecules like proteins, DNA, mRNA, lipids, and miRNA. These vesicles are abundant in fluids distributed throughout the body, including urine, blood, saliva, and even bile. They are important diagnostic tools for breast, lung, gastrointestinal cancers, etc. However, their application as cancer biomarkers has not yet been implemented in most parts of the world. In this review, we discuss how OMICs profiling of exosomes can be practiced by substituting traditional imaging or biopsy methods for cancer detection. Previous methods like extensive imaging and biopsy used for screening were expensive, mostly invasive, and could not easily provide early detection for various types of cancer. Exosomal biomarkers can be utilized for routine screening by simply collecting body fluids from the individual. We anticipate that the use of exosomes will be brought to light by the success of clinical trials investigating their potential to enhance cancer detection and treatment in the upcoming years.

众所周知,外泌体是一种细胞外囊泡,可通过分布蛋白质、DNA、mRNA、脂质和 miRNA 等重要大分子促进细胞间通信。这些囊泡大量存在于尿液、血液、唾液甚至胆汁等全身液体中。它们是乳腺癌、肺癌、胃肠道癌症等的重要诊断工具。然而,它们作为癌症生物标志物的应用尚未在世界大部分地区开展。在这篇综述中,我们将讨论如何通过外泌体的 OMICs 分析来替代传统的成像或活检方法,从而进行癌症检测。以前用于筛查的广泛成像和活组织检查等方法费用昂贵,大多为侵入性检查,而且不容易对各种类型的癌症进行早期检测。外泌体生物标志物可用于常规筛查,只需采集个人体液即可。我们预计,在未来几年里,外泌体在加强癌症检测和治疗方面的潜力将随着临床试验的成功而得到广泛应用。
{"title":"Harnessing exosomes as cancer biomarkers in clinical oncology.","authors":"Subhrojyoti Ghosh, Ramya Lakshmi Rajendran, Atharva A Mahajan, Ankita Chowdhury, Aishi Bera, Sudeepta Guha, Kashmira Chakraborty, Rajanyaa Chowdhury, Aritra Paul, Shreya Jha, Anuvab Dey, Amit Dubey, Sukhamoy Gorai, Purbasha Das, Chae Moon Hong, Anand Krishnan, Prakash Gangadaran, Byeong-Cheol Ahn","doi":"10.1186/s12935-024-03464-5","DOIUrl":"10.1186/s12935-024-03464-5","url":null,"abstract":"<p><p>Exosomes are extracellular vesicles well known for facilitating cell-to-cell communication by distributing essential macromolecules like proteins, DNA, mRNA, lipids, and miRNA. These vesicles are abundant in fluids distributed throughout the body, including urine, blood, saliva, and even bile. They are important diagnostic tools for breast, lung, gastrointestinal cancers, etc. However, their application as cancer biomarkers has not yet been implemented in most parts of the world. In this review, we discuss how OMICs profiling of exosomes can be practiced by substituting traditional imaging or biopsy methods for cancer detection. Previous methods like extensive imaging and biopsy used for screening were expensive, mostly invasive, and could not easily provide early detection for various types of cancer. Exosomal biomarkers can be utilized for routine screening by simply collecting body fluids from the individual. We anticipate that the use of exosomes will be brought to light by the success of clinical trials investigating their potential to enhance cancer detection and treatment in the upcoming years.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-08-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11308730/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141901030","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Enhanced anti-tumor effects by combination of tucatinib and radiation in HER2-overexpressing human cancer cell lines. 图卡替尼和辐射联合使用可增强对HER2表达的人类癌细胞株的抗肿瘤效果。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-06 DOI: 10.1186/s12935-024-03458-3
Lukas Amrell, Eric Bär, Annegret Glasow, Rolf-Dieter Kortmann, Clemens Seidel, Ina Patties

Background: Tucatinib (TUC), a HER2-directed tyrosine kinase inhibitor, is the first targeted drug demonstrating intracranial efficacy and significantly prolonged survival in metastatic HER2-positive breast cancer (BC) patients with brain metastases. Current treatments for brain metastases often include radiotherapy, but little is known about the effects of combination treatment with TUC. Therefore, we examined the combined effects of irradiation and TUC in human HER2-overexpressing BC, non-small cell lung cancer (NSCLC), and colorectal cancer (CRC) cell lines. For the latter two, a standard therapy successfully targeting HER2 is yet to be established.

Methods: Nine HER2-overexpressing (BC: BT474, ZR7530, HCC1954; CRC: LS411N, DLD1, COLO201; NSCLC: DV90, NCI-H1781) and three control cell lines (BC: MCF7, HCC38; NSCLC: NCI-H2030) were examined. WST-1 assay (metabolic activity), BrdU ELISA (proliferation), γH2AX assay (DNA double-strand breaks (DSB), Annexin V assay (apoptosis), and clonogenic assay (clonogenicity) were performed after treatment with TUC and/or irradiation (IR). The relevance of the treatment sequence was analyzed exemplarily.

Results: In BC, combinatorial treatment with TUC and IR significantly decreased metabolic activity, cell proliferation, clonogenicity and enhanced apoptotis compared to IR alone, whereby cell line-specific differences occurred. In the PI3KCA-mutated HCC1954 cell line, addition of alpelisib (ALP) further decreased clonogenicity. TUC delayed the repair of IR-induced DNA damage but did not induce DSB itself. Investigation of treatment sequence indicated a benefit of IR before TUC versus IR after TUC. Also in CRC and NSCLC, the combination led to a stronger inhibition of metabolic activity, proliferation, and clonogenic survival (only in NSCLC) than IR alone, whereby about 10-fold higher concentrations of TUC had to be applied than in BC to induce significant changes.

Conclusion: Our data indicate that combination of TUC and IR could be more effective than single treatment strategies for BC. Thereby, treatment sequence seems to be an important factor. The lower sensitivity to TUC in NSCLC and particularly in CRC (compared to BC) implicates, that tumor promotion there might be less HER2-related. Combination with inhibitors of other driver mutations may aid in overcoming partial TUC resistance. These findings are of high relevance to improve long-time prognosis especially in brain-metastasized situations given the intracranial activity of TUC.

背景:图卡替尼(TUC)是一种HER2导向的酪氨酸激酶抑制剂,它是首个显示出颅内疗效的靶向药物,可显著延长HER2阳性转移性乳腺癌(BC)脑转移患者的生存期。目前治疗脑转移的方法通常包括放疗,但对与 TUC 联合治疗的效果知之甚少。因此,我们在人类 HER2 表达阳性 BC、非小细胞肺癌(NSCLC)和结直肠癌(CRC)细胞系中研究了照射和 TUC 的联合效果。对于后两者,成功靶向 HER2 的标准疗法尚未确立:方法:九种 HER2 基因表达过高的细胞系(BC:BT474, ZR7530, HCC1954; CRC: ls411n, Dld1, colo201; NSCLC:DV90、NCI-H1781)和三个对照细胞系(BC:MCF7、HCC38;NSCLC:NCI-H2030)进行了研究。在使用 TUC 和/或辐照(IR)处理后,进行了 WST-1 检测(代谢活性)、BrdU ELISA 检测(增殖)、γH2AX 检测(DNA 双链断裂(DSB))、Annexin Vay 检测(细胞凋亡)和克隆生成检测(克隆生成性)。结果表明:在 BC 中,TUC 和/或照射(IR)的组合治疗可抑制细胞凋亡:结果:与单独使用IR相比,在BC中使用TUC和IR联合治疗可显著降低代谢活性、细胞增殖、克隆生成和增强细胞凋亡,其中存在细胞系特异性差异。在PI3KCA突变的HCC1954细胞系中,添加阿来替尼(ALP)可进一步降低细胞的克隆性。TUC 可延缓红外诱导的 DNA 损伤的修复,但本身不会诱导 DSB。对治疗顺序的研究表明,在 TUC 之前进行红外治疗比在 TUC 之后进行红外治疗更有利。在 CRC 和 NSCLC 中,联合用药对代谢活性、增殖和克隆存活(仅在 NSCLC 中)的抑制作用比单独使用 IR 更强,其中 TUC 的浓度必须比 BC 高约 10 倍才能诱发显著变化:我们的数据表明,TUC 和 IR 联合治疗 BC 比单一治疗策略更有效。因此,治疗顺序似乎是一个重要因素。NSCLC尤其是CRC(与BC相比)对TUC的敏感性较低,这说明肿瘤的生长可能与HER2的关系不大。与其他驱动突变抑制剂联合使用可能有助于克服TUC的部分耐药性。鉴于 TUC 在颅内的活性,这些发现对于改善长期预后,尤其是脑转移情况下的预后具有重要意义。
{"title":"Enhanced anti-tumor effects by combination of tucatinib and radiation in HER2-overexpressing human cancer cell lines.","authors":"Lukas Amrell, Eric Bär, Annegret Glasow, Rolf-Dieter Kortmann, Clemens Seidel, Ina Patties","doi":"10.1186/s12935-024-03458-3","DOIUrl":"10.1186/s12935-024-03458-3","url":null,"abstract":"<p><strong>Background: </strong>Tucatinib (TUC), a HER2-directed tyrosine kinase inhibitor, is the first targeted drug demonstrating intracranial efficacy and significantly prolonged survival in metastatic HER2-positive breast cancer (BC) patients with brain metastases. Current treatments for brain metastases often include radiotherapy, but little is known about the effects of combination treatment with TUC. Therefore, we examined the combined effects of irradiation and TUC in human HER2-overexpressing BC, non-small cell lung cancer (NSCLC), and colorectal cancer (CRC) cell lines. For the latter two, a standard therapy successfully targeting HER2 is yet to be established.</p><p><strong>Methods: </strong>Nine HER2-overexpressing (BC: BT474, ZR7530, HCC1954; CRC: LS411N, DLD1, COLO201; NSCLC: DV90, NCI-H1781) and three control cell lines (BC: MCF7, HCC38; NSCLC: NCI-H2030) were examined. WST-1 assay (metabolic activity), BrdU ELISA (proliferation), γH2AX assay (DNA double-strand breaks (DSB), Annexin V assay (apoptosis), and clonogenic assay (clonogenicity) were performed after treatment with TUC and/or irradiation (IR). The relevance of the treatment sequence was analyzed exemplarily.</p><p><strong>Results: </strong>In BC, combinatorial treatment with TUC and IR significantly decreased metabolic activity, cell proliferation, clonogenicity and enhanced apoptotis compared to IR alone, whereby cell line-specific differences occurred. In the PI3KCA-mutated HCC1954 cell line, addition of alpelisib (ALP) further decreased clonogenicity. TUC delayed the repair of IR-induced DNA damage but did not induce DSB itself. Investigation of treatment sequence indicated a benefit of IR before TUC versus IR after TUC. Also in CRC and NSCLC, the combination led to a stronger inhibition of metabolic activity, proliferation, and clonogenic survival (only in NSCLC) than IR alone, whereby about 10-fold higher concentrations of TUC had to be applied than in BC to induce significant changes.</p><p><strong>Conclusion: </strong>Our data indicate that combination of TUC and IR could be more effective than single treatment strategies for BC. Thereby, treatment sequence seems to be an important factor. The lower sensitivity to TUC in NSCLC and particularly in CRC (compared to BC) implicates, that tumor promotion there might be less HER2-related. Combination with inhibitors of other driver mutations may aid in overcoming partial TUC resistance. These findings are of high relevance to improve long-time prognosis especially in brain-metastasized situations given the intracranial activity of TUC.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11302197/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141896772","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
期刊
Cancer Cell International
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