首页 > 最新文献

中国肺癌杂志最新文献

英文 中文
[A Case Report of EGFR-TKIs Resistant Secondary MET Gene Amplified 
Lung Squamous Cell Carcinoma and Literature Review]. [EGFR-TKIs耐药继发性MET基因扩增1例报道
肺鳞癌及文献复习]。
Q4 Medicine Pub Date : 2024-11-20 DOI: 10.3779/j.issn.1009-3419.2024.106.32
Yalan Liu, Peng Chen, Xinfu Liu

With the rapid development of epidermal growth factor receptor (EGFR) gene testing of lung adenocarcinoma patients has been routinely carried out, EGFR mutations are also possible for some small samples of non-smoking female lung squamous cell carcinoma patients. This increases the opportunity for targeted therapy for this group of patients. However, drug resistance in patients with lung squamous cell carcinoma during targeted therapy is an important factor affecting subsequent treatment. There are multiple mechanisms of acquired drug resistance in targeted therapy, and the alteration of mesenchymal-epithelial transition factor (MET) signaling pathway is one of the common mechanisms of drug resistance. At present, some selective tyrosine kinase inhibitors (TKIs) of MET has been approved for non-small cell lung cancer with MET gene 14 exon skipping mutation, such as Glumetinib, Savolitinib, Tepotinib, Capmatinib, etc. Drugs that target secondary MET amplification are still in clinical trials. This paper retrospectively analyzed the clinical data of a female patient with EGFR-TKIs resistant secondary MET amplified squamous cell lung cancer, and reviewed relevant literature to explore how to optimize the treatment of lung squamous cell carcinoma patients with EGFR mutation, so as to provide clinical reference for the diagnosis and treatment of such patients.
.

随着肺腺癌患者表皮生长因子受体(epidermal growth factor receptor, EGFR)基因检测的快速发展,一些小样本非吸烟女性肺鳞状细胞癌患者也可能出现EGFR突变。这增加了对这组患者进行靶向治疗的机会。然而,肺鳞状细胞癌患者在靶向治疗过程中的耐药是影响后续治疗的重要因素。靶向治疗的获得性耐药机制多种多样,间充质-上皮过渡因子(mesenchymal-epithelial transition factor, MET)信号通路改变是常见的耐药机制之一。目前,MET的一些选择性酪氨酸激酶抑制剂(TKIs)已被批准用于MET基因14外显子跳变的非小细胞肺癌,如Glumetinib、Savolitinib、tepo替尼、Capmatinib等。针对二次MET扩增的药物仍处于临床试验阶段。本文回顾性分析1例女性EGFR- tkis耐药继发性MET扩增型鳞状细胞肺癌患者的临床资料,并复习相关文献,探讨如何优化EGFR突变型肺鳞状细胞癌患者的治疗,为该类患者的诊治提供临床参考。
{"title":"[A Case Report of EGFR-TKIs Resistant Secondary MET Gene Amplified \u2029Lung Squamous Cell Carcinoma and Literature Review].","authors":"Yalan Liu, Peng Chen, Xinfu Liu","doi":"10.3779/j.issn.1009-3419.2024.106.32","DOIUrl":"10.3779/j.issn.1009-3419.2024.106.32","url":null,"abstract":"<p><p>With the rapid development of epidermal growth factor receptor (EGFR) gene testing of lung adenocarcinoma patients has been routinely carried out, EGFR mutations are also possible for some small samples of non-smoking female lung squamous cell carcinoma patients. This increases the opportunity for targeted therapy for this group of patients. However, drug resistance in patients with lung squamous cell carcinoma during targeted therapy is an important factor affecting subsequent treatment. There are multiple mechanisms of acquired drug resistance in targeted therapy, and the alteration of mesenchymal-epithelial transition factor (MET) signaling pathway is one of the common mechanisms of drug resistance. At present, some selective tyrosine kinase inhibitors (TKIs) of MET has been approved for non-small cell lung cancer with MET gene 14 exon skipping mutation, such as Glumetinib, Savolitinib, Tepotinib, Capmatinib, etc. Drugs that target secondary MET amplification are still in clinical trials. This paper retrospectively analyzed the clinical data of a female patient with EGFR-TKIs resistant secondary MET amplified squamous cell lung cancer, and reviewed relevant literature to explore how to optimize the treatment of lung squamous cell carcinoma patients with EGFR mutation, so as to provide clinical reference for the diagnosis and treatment of such patients.\u2029.</p>","PeriodicalId":39317,"journal":{"name":"中国肺癌杂志","volume":"27 11","pages":"878-884"},"PeriodicalIF":0.0,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732385/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972551","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
[Application of Nano-drug Delivery Technology in Overcoming Drug Resistance 
in Lung Cancer]. [纳米给药技术在克服肺癌耐药中的应用
]。
Q4 Medicine Pub Date : 2024-11-20 DOI: 10.3779/j.issn.1009-3419.2024.101.30
Yingchun Lu, Chunyu Wang, Bin Liu

Lung cancer is one of the most malignant tumor, representing a significant threat to human health. In China, its mortality rate is the highest among all malignant tumors. The occurrence of drug resistance has resulted in unfavourable prognosis for patients with lung cancer, and overcoming drug resistance is a significant challenge that needs to be addressed. Nano-drug delivery technology has been an important approach to overcome drug resistance in lung cancer. Targeting to the mechanisms of drug resistance, by enabling the combined delivery of drugs, increasing the efficiency of drug delivery and improving the targeting and safety of drugs, nano-drug delivery technology offers a novel approach to tackling drug resistance in lung cancer. This paper describes the current status of lung cancer treatment, mechanisms of drug resistance, strategies to overcome drug resistance, and the application of nanotechnology in the diagnosis and treatment of lung cancer. In addition, it summarizes the recent research progress on the application of nano-drug delivery technology to overcome drug resistance in lung cancer. Finally, the current prospects and challenges of nano-drug delivery technology are discussed.
.

肺癌是最恶性的肿瘤之一,严重威胁着人类的健康。在中国,其死亡率是所有恶性肿瘤中最高的。耐药的发生导致肺癌患者预后不利,克服耐药是一个需要解决的重大挑战。纳米给药技术已成为克服肺癌耐药的重要途径。纳米给药技术针对耐药机制,通过联合给药,提高给药效率,改善药物的靶向性和安全性,为解决肺癌耐药问题提供了新的途径。本文介绍了肺癌的治疗现状、耐药机制、克服耐药的策略以及纳米技术在肺癌诊断和治疗中的应用。此外,综述了纳米给药技术在克服肺癌耐药中的应用研究进展。最后,对纳米给药技术的发展前景和面临的挑战进行了讨论。
。
{"title":"[Application of Nano-drug Delivery Technology in Overcoming Drug Resistance \u2029in Lung Cancer].","authors":"Yingchun Lu, Chunyu Wang, Bin Liu","doi":"10.3779/j.issn.1009-3419.2024.101.30","DOIUrl":"10.3779/j.issn.1009-3419.2024.101.30","url":null,"abstract":"<p><p>Lung cancer is one of the most malignant tumor, representing a significant threat to human health. In China, its mortality rate is the highest among all malignant tumors. The occurrence of drug resistance has resulted in unfavourable prognosis for patients with lung cancer, and overcoming drug resistance is a significant challenge that needs to be addressed. Nano-drug delivery technology has been an important approach to overcome drug resistance in lung cancer. Targeting to the mechanisms of drug resistance, by enabling the combined delivery of drugs, increasing the efficiency of drug delivery and improving the targeting and safety of drugs, nano-drug delivery technology offers a novel approach to tackling drug resistance in lung cancer. This paper describes the current status of lung cancer treatment, mechanisms of drug resistance, strategies to overcome drug resistance, and the application of nanotechnology in the diagnosis and treatment of lung cancer. In addition, it summarizes the recent research progress on the application of nano-drug delivery technology to overcome drug resistance in lung cancer. Finally, the current prospects and challenges of nano-drug delivery technology are discussed.\u2029.</p>","PeriodicalId":39317,"journal":{"name":"中国肺癌杂志","volume":"27 11","pages":"864-872"},"PeriodicalIF":0.0,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732387/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972555","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
[Savolitinib Induced Pathological Complete Response in Non-small Cell Lung Cancer with MET Amplification: A Case Report].
Q4 Medicine Pub Date : 2024-11-20 DOI: 10.3779/j.issn.1009-3419.2024.102.36
Meng Lu, Ran Zhang, Baiwei Li, Haidi Xu, Yongkuan Guo, Jian You, Bingsheng Sun

Mesenchymal-epithelial transition factor (MET) gene mutation is a large class of mutations commonly seen in non-small cell lung cancer (NSCLC). MET mutation includes subtypes such as MET exon 14 skipping mutation (METex14m) and MET amplification (METamp). For advanced NSCLC with METex14m, Savolitinib has a high sensitivity as a member of tyrosine kinase inhibitors (TKIs). METamp is a relatively rare genetic mutation type which can serve as a driver gene to mediate primary and later acquired drug resistance of epidermal growth factor receptor (EGFR)-TKIs. For advanced NSCLC with secondary METamp, EGFR-TKIs combined with MET-TKIs are usually used in clinical treatment, while the optimal treatment strategy for advanced NSCLC with primary METamp has not yet been determined. For locally advanced NSCLC patients with positive driver gene mutations such as EGFR, anaplastic lymphoma kinase (ALK) fusion and METex14m, there have been relevant cases reported that neoadjuvant targeted therapy could achieve a good prognosis, but there have been no cases of neoadjuvant targeted therapy for locally advanced NSCLC patients with METamp. This report describes a case of a locally advanced NSCLC patient with dual driver gene mutations (EGFR L858R combined with primary METamp), the tumor did not shrink after 1 month of Gefitinib monotherapy, but significantly subsided after 4 months of Savolitinib monotherapy. After radical surgery, the pathological results proved pathological complete response (pCR) of the tumor, and the patient had a good response to postoperative continual Savolitinib treatment, with no recurrence nor metastasis observed to date. This case reports the feasibility and effectiveness of neoadjuvant targeted therapy for locally advanced NSCLC with primary METamp, aiming to provide effective reference for perioperative treatment of locally advanced NSCLC with primary METamp.
.

{"title":"[Savolitinib Induced Pathological Complete Response in Non-small Cell Lung Cancer with MET Amplification: A Case Report].","authors":"Meng Lu, Ran Zhang, Baiwei Li, Haidi Xu, Yongkuan Guo, Jian You, Bingsheng Sun","doi":"10.3779/j.issn.1009-3419.2024.102.36","DOIUrl":"https://doi.org/10.3779/j.issn.1009-3419.2024.102.36","url":null,"abstract":"<p><p>Mesenchymal-epithelial transition factor (MET) gene mutation is a large class of mutations commonly seen in non-small cell lung cancer (NSCLC). MET mutation includes subtypes such as MET exon 14 skipping mutation (METex14m) and MET amplification (METamp). For advanced NSCLC with METex14m, Savolitinib has a high sensitivity as a member of tyrosine kinase inhibitors (TKIs). METamp is a relatively rare genetic mutation type which can serve as a driver gene to mediate primary and later acquired drug resistance of epidermal growth factor receptor (EGFR)-TKIs. For advanced NSCLC with secondary METamp, EGFR-TKIs combined with MET-TKIs are usually used in clinical treatment, while the optimal treatment strategy for advanced NSCLC with primary METamp has not yet been determined. For locally advanced NSCLC patients with positive driver gene mutations such as EGFR, anaplastic lymphoma kinase (ALK) fusion and METex14m, there have been relevant cases reported that neoadjuvant targeted therapy could achieve a good prognosis, but there have been no cases of neoadjuvant targeted therapy for locally advanced NSCLC patients with METamp. This report describes a case of a locally advanced NSCLC patient with dual driver gene mutations (EGFR L858R combined with primary METamp), the tumor did not shrink after 1 month of Gefitinib monotherapy, but significantly subsided after 4 months of Savolitinib monotherapy. After radical surgery, the pathological results proved pathological complete response (pCR) of the tumor, and the patient had a good response to postoperative continual Savolitinib treatment, with no recurrence nor metastasis observed to date. This case reports the feasibility and effectiveness of neoadjuvant targeted therapy for locally advanced NSCLC with primary METamp, aiming to provide effective reference for perioperative treatment of locally advanced NSCLC with primary METamp.\u2029.</p>","PeriodicalId":39317,"journal":{"name":"中国肺癌杂志","volume":"27 11","pages":"873-877"},"PeriodicalIF":0.0,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732386/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143056106","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
[Immunotherapy for Extensive-stage Small Cell Lung Cancer: 
Research Progress and Future Perspectives]. [广泛期小细胞肺癌的免疫治疗:
研究进展和未来展望]。
Q4 Medicine Pub Date : 2024-11-20 DOI: 10.3779/j.issn.1009-3419.2024.102.35
Yuling Zhong, Jingyi Wang, Lin Wu

At present, immunotherapy combined with chemotherapy has become the first-line standard of treatment for extensive-stage small cell lung cancer (ES-SCLC). In recent years, immune checkpoint inhibitors (ICIs) have received extensive attention and research in the field of lung cancer. At the same time, there are many challenges and tests in this process, such as the exploration of biomarkers, the exploration of new targets and new models, and the management of special populations. This article reviews the research progress in the field of ES-SCLC immunotherapy, and looks forward to the future development trend and potential direction of this field.
.

目前,免疫联合化疗已成为广泛期小细胞肺癌(ES-SCLC)的一线治疗标准。近年来,免疫检查点抑制剂(ICIs)在肺癌领域得到了广泛的关注和研究。同时,在这一过程中也面临着诸多挑战和考验,如生物标志物的探索、新靶点和新模式的探索、特殊人群的管理等。本文综述了ES-SCLC免疫治疗领域的研究进展,并展望了该领域未来的发展趋势和潜在方向。
。
{"title":"[Immunotherapy for Extensive-stage Small Cell Lung Cancer: \u2029Research Progress and Future Perspectives].","authors":"Yuling Zhong, Jingyi Wang, Lin Wu","doi":"10.3779/j.issn.1009-3419.2024.102.35","DOIUrl":"10.3779/j.issn.1009-3419.2024.102.35","url":null,"abstract":"<p><p>At present, immunotherapy combined with chemotherapy has become the first-line standard of treatment for extensive-stage small cell lung cancer (ES-SCLC). In recent years, immune checkpoint inhibitors (ICIs) have received extensive attention and research in the field of lung cancer. At the same time, there are many challenges and tests in this process, such as the exploration of biomarkers, the exploration of new targets and new models, and the management of special populations. This article reviews the research progress in the field of ES-SCLC immunotherapy, and looks forward to the future development trend and potential direction of this field.\u2029.</p>","PeriodicalId":39317,"journal":{"name":"中国肺癌杂志","volume":"27 11","pages":"855-863"},"PeriodicalIF":0.0,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732389/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972559","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
[Non-small Cell Lung Cancer Cell Line PC-9 Drug-resistant Mutant Cell Line 
Establishment and Validation of Their Sensitivity to EGFR Inhibitors]. [非小细胞肺癌细胞系PC-9耐药突变细胞系
对EGFR抑制剂敏感性的建立及验证]。
Q4 Medicine Pub Date : 2024-11-20 DOI: 10.3779/j.issn.1009-3419.2024.101.31
Tao Hu, Yang Lou, Mingbo Su
<p><strong>Background: </strong>Mutations in the structural domain of the epidermal growth factor receptor (EGFR) kinase represent a critical pathogenetic factor in non-small cell lung cancer (NSCLC). Small-molecule EGFR-tyrosine kinase inhibitors (TKIs) serve as first-line therapeutic agents for the treatment of EGFR-mutated NSCLC. But the resistance mutations of EGFR restrict the clinical application of EGFR-TKIs. In this study, we constructed a clinically relevant PC-9 EGFRD19/T790M/C797S cellular model featuring the mutation type within the EGFRD19/T790M/C797S. This model aims to investigate the inhibitory effects of small-molecule EGFR-TKIs and to provide a cellular platform for developing a new generation of innovative drugs that target resistance associated with EGFR mutations.</p><p><strong>Methods: </strong>Clustered regularly interspaced short palindromic repeats/CRISPR-associated nuclease 9 (CRISPR/Cas9) technology was employed to knock in the EGFRT790M/C797S mutant fragment into NSCLC PC-9 cells, originally harboring the EGFRD19 mutation, to generate the PC-9 EGFRD19/T790M/C797S cell model. This model, with the EGFRD19/T790M/C797S mutant, was used to investigate the inhibitory effects of EGFR-TKIs on cell proliferation through MTS assay. Additionally, Western blot analysis was conducted to assess the regulation of EGFR protein expression and the phosphorylation levels of downstream signaling molecules, including protein kinase B (AKT) and mitogen-activated protein kinase (MAPK).</p><p><strong>Results: </strong>PC-9 EGFRD19/T790M/C797S cells, with the EGFRD19/T790M/C797S mutation, were successfully generated using CRISPR/Cas9 technology. In terms of proliferation inhibition, the marketed first-, second-, and third-generation EGFR-TKIs that were ineffective against the EGFRD19/T790M/C797S mutation showed weak proliferation inhibitory activity against this cell line, and the proliferation inhibition (half maximal inhibitory concentration, IC50)>1000 nmol/L; in contrast, the fourth-generation EGFR-TKIs in development, which have better efficacy against the EGFRD19/T790M/C797S mutation, showed strong proliferation inhibition in this cell model. On mechanistic validation, the first-, second-, and third-generation EGFR-TKIs had weak inhibitory activity on the phosphorylation of EGFR and the downstream AKT/MAPK signaling pathway in this cell line, whereas the fourth generation of EGFR-TKIs under development significantly inhibited the phosphorylation of EGFR and the downstream AKT/MAPK signaling pathway in this cell line.</p><p><strong>Conclusions: </strong>Using CRISPR/Cas9 technology, the EGFRT790M/C797S mutant fragment was successfully knocked into PC-9 cells to create cell lines harboring the EGFRD19/T790M/C797S mutation. The study demonstrated that the EGFR-TKIs showed different sensitivities to whether the EGFRD19/T790M/C797S mutation was effective or not and different inhibitory effects on the phosphorylation of EGFR and downstream pat
背景:表皮生长因子受体(EGFR)激酶结构域的突变是非小细胞肺癌(NSCLC)的一个关键致病因素。小分子egfr -酪氨酸激酶抑制剂(TKIs)是治疗egfr突变的非小细胞肺癌的一线治疗药物。但EGFR的耐药突变限制了EGFR- tkis的临床应用。在本研究中,我们构建了具有临床相关性的PC-9 EGFRD19/T790M/C797S细胞模型,该模型具有EGFRD19/T790M/C797S内的突变类型。该模型旨在研究小分子EGFR- tkis的抑制作用,并为开发新一代靶向与EGFR突变相关的耐药性的创新药物提供细胞平台。方法:采用聚类规则间隔短回词重复/CRISPR相关核酸酶9 (CRISPR/Cas9)技术,将EGFRT790M/C797S突变片段敲入原含有EGFRD19突变的NSCLC PC-9细胞中,生成PC-9 EGFRD19/T790M/C797S细胞模型。该模型采用EGFRD19/T790M/C797S突变体,通过MTS法研究EGFR-TKIs对细胞增殖的抑制作用。此外,我们还进行了Western blot分析,以评估EGFR蛋白表达的调控以及下游信号分子的磷酸化水平,包括蛋白激酶B (AKT)和丝裂原活化蛋白激酶(MAPK)。结果:利用CRISPR/Cas9技术成功生成EGFRD19/T790M/C797S突变的PC-9 EGFRD19/T790M/C797S细胞在增殖抑制方面,已上市的第一代、第二代和第三代EGFR-TKIs对EGFRD19/T790M/C797S突变无效,对该细胞系的增殖抑制活性较弱,增殖抑制(最大抑制浓度的一半,IC50)低于1000 nmol/L;相比之下,正在开发的第四代EGFR-TKIs对EGFRD19/T790M/C797S突变有更好的疗效,在该细胞模型中表现出较强的增殖抑制作用。在机制验证中,第一代、第二代和第三代EGFR- tkis在该细胞系中对EGFR磷酸化和下游AKT/MAPK信号通路具有较弱的抑制活性,而正在开发的第四代EGFR- tkis在该细胞系中显著抑制EGFR磷酸化和下游AKT/MAPK信号通路。结论:利用CRISPR/Cas9技术,EGFRT790M/C797S突变片段成功敲入PC-9细胞,构建了EGFRD19/T790M/C797S突变细胞系。研究表明,EGFR- tkis对EGFRD19/T790M/C797S突变是否有效表现出不同的敏感性,对EGFR磷酸化及下游通路的抑制作用也不同,表明该细胞系依赖于EGFRD19/T790M/C797S突变和EGFR/AKT/MAPK信号通路的激活进行增殖。本研究为开发新一代靶向EGFR突变耐药的创新药物提供了临床相关的细胞评价和机制验证体系。
{"title":"[Non-small Cell Lung Cancer Cell Line PC-9 Drug-resistant Mutant Cell Line \u2029Establishment and Validation of Their Sensitivity to EGFR Inhibitors].","authors":"Tao Hu, Yang Lou, Mingbo Su","doi":"10.3779/j.issn.1009-3419.2024.101.31","DOIUrl":"10.3779/j.issn.1009-3419.2024.101.31","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Mutations in the structural domain of the epidermal growth factor receptor (EGFR) kinase represent a critical pathogenetic factor in non-small cell lung cancer (NSCLC). Small-molecule EGFR-tyrosine kinase inhibitors (TKIs) serve as first-line therapeutic agents for the treatment of EGFR-mutated NSCLC. But the resistance mutations of EGFR restrict the clinical application of EGFR-TKIs. In this study, we constructed a clinically relevant PC-9 EGFRD19/T790M/C797S cellular model featuring the mutation type within the EGFRD19/T790M/C797S. This model aims to investigate the inhibitory effects of small-molecule EGFR-TKIs and to provide a cellular platform for developing a new generation of innovative drugs that target resistance associated with EGFR mutations.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;Clustered regularly interspaced short palindromic repeats/CRISPR-associated nuclease 9 (CRISPR/Cas9) technology was employed to knock in the EGFRT790M/C797S mutant fragment into NSCLC PC-9 cells, originally harboring the EGFRD19 mutation, to generate the PC-9 EGFRD19/T790M/C797S cell model. This model, with the EGFRD19/T790M/C797S mutant, was used to investigate the inhibitory effects of EGFR-TKIs on cell proliferation through MTS assay. Additionally, Western blot analysis was conducted to assess the regulation of EGFR protein expression and the phosphorylation levels of downstream signaling molecules, including protein kinase B (AKT) and mitogen-activated protein kinase (MAPK).&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;PC-9 EGFRD19/T790M/C797S cells, with the EGFRD19/T790M/C797S mutation, were successfully generated using CRISPR/Cas9 technology. In terms of proliferation inhibition, the marketed first-, second-, and third-generation EGFR-TKIs that were ineffective against the EGFRD19/T790M/C797S mutation showed weak proliferation inhibitory activity against this cell line, and the proliferation inhibition (half maximal inhibitory concentration, IC50)&gt;1000 nmol/L; in contrast, the fourth-generation EGFR-TKIs in development, which have better efficacy against the EGFRD19/T790M/C797S mutation, showed strong proliferation inhibition in this cell model. On mechanistic validation, the first-, second-, and third-generation EGFR-TKIs had weak inhibitory activity on the phosphorylation of EGFR and the downstream AKT/MAPK signaling pathway in this cell line, whereas the fourth generation of EGFR-TKIs under development significantly inhibited the phosphorylation of EGFR and the downstream AKT/MAPK signaling pathway in this cell line.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusions: &lt;/strong&gt;Using CRISPR/Cas9 technology, the EGFRT790M/C797S mutant fragment was successfully knocked into PC-9 cells to create cell lines harboring the EGFRD19/T790M/C797S mutation. The study demonstrated that the EGFR-TKIs showed different sensitivities to whether the EGFRD19/T790M/C797S mutation was effective or not and different inhibitory effects on the phosphorylation of EGFR and downstream pat","PeriodicalId":39317,"journal":{"name":"中国肺癌杂志","volume":"27 11","pages":"815-825"},"PeriodicalIF":0.0,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732384/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972568","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
[Clinicopathological Analysis of 14 Cases of Primary Pulmonary Lymphoepithelial Carcinoma]. 原发性肺淋巴上皮癌14例临床病理分析
Q4 Medicine Pub Date : 2024-11-20 DOI: 10.3779/j.issn.1009-3419.2024.101.29
Yixuan Fang, Anzhe Wang, Lumin Shen, Xiao Yuan, Yu Kong

Background: Primary pulmonary lymphoepithelial carcinoma (PPLEC) is a rare form of lung malignancy, accounting for only 0.7% of all lung cancers. It is currently classified as a distinct subtype within squamous cell carcinomas. This study aims to explore the clinicopathological characteristics of PPLEC and its subtypes, with the objective of enhancing understanding and improving diagnostic accuracy for this disease.

Methods: A retrospective analysis was conducted on the clinical, pathological, imaging, and prognostic data of 14 patients diagnosed with PPLEC at the First Affiliated Hospital of Soochow University between February 2019 and June 2023.

Results: A total of 14 cases of PPLEC were identified, including 5 cases of the Regaud type, with ages ranging from 33 to 73 years, comprising 2 males and 3 females; and 9 cases of the Schmincke type, with ages ranging from 36 to 79 years, including 4 males and 5 females. Computed tomography (CT) scans consistently demonstrated soft tissue masses or nodular shadows. Reagud type mainly showed peripheral masses and Schmincke type mainly showed central masses. Pathological examination revealed tumor cells exhibiting syncytial-like growth, accompanied by lymphocytic infiltration and stromal fibrosis, with the Regaud type showing well-defined borders combined with granulomatous inflammation, while the Schmincke type exhibited indistinct tumor margins. Immunohistochemistry showed that CK, CK5/6, P40 and P63 were positive, and the Ki-67 index of Regaud type was lower than that of Schmincke type; notably, all 8 cases tested for programmed death-ligand 1 (PD-L1) were positive. Epstein-Barr virus-encoded RNA (EBER) in situ hybridization was positive in all instances. Among these cases, 6 underwent surgical treatment, and 8 received comprehensive therapy; by the end of the follow-up period, all 14 patients remained alive.

Conclusions: PPLEC is a rare form of malignant lung tumor associated with Epstein-Barr virus (EBV) infection. The Regaud and Schmincke subtypes display distinct imaging and pathological characteristics. In the early stages of the disease, surgical intervention is the primary treatment method; however, for advanced stages, a multimodal treatment approach is utilized, resulting in a relatively favorable prognosis. Immunotherapy represents a promising and effective treatment modality for patients with middle to advanced stage disease exhibiting high PD-L1 expression levels.

背景:原发性肺淋巴上皮癌(PPLEC)是一种罕见的肺部恶性肿瘤,仅占所有肺癌的0.7%。它目前被归类为鳞状细胞癌中一个独特的亚型。本研究旨在探讨PPLEC及其亚型的临床病理特征,以提高对该病的认识和诊断准确性。方法:回顾性分析2019年2月至2023年6月在苏州大学第一附属医院诊断为PPLEC的14例患者的临床、病理、影像学和预后资料。结果:共发现PPLEC 14例,其中Regaud型5例,年龄33 ~ 73岁,男2例,女3例;Schmincke型9例,年龄36 ~ 79岁,男4例,女5例。计算机断层扫描(CT)一致显示软组织肿块或结节影。Reagud型以周围团块为主,Schmincke型以中心团块为主。病理检查显示肿瘤细胞表现为合胞样生长,伴淋巴细胞浸润及间质纤维化,Regaud型边界清晰并伴有肉芽肿性炎症,Schmincke型肿瘤边缘不清。免疫组化示CK、CK5/6、P40、P63阳性,且Regaud型Ki-67指数低于Schmincke型;值得注意的是,所有8例程序性死亡配体1 (PD-L1)检测均呈阳性。Epstein-Barr病毒编码RNA (EBER)原位杂交在所有病例中均为阳性。其中手术治疗6例,综合治疗8例;到随访期结束时,所有14名患者仍然存活。结论:PPLEC是一种罕见的与eb病毒(EBV)感染相关的恶性肺肿瘤。Regaud和Schmincke亚型表现出不同的影像学和病理特征。在疾病早期,手术干预是主要的治疗方法;然而,对于晚期,采用多模式治疗方法,预后相对较好。对于PD-L1高表达的中晚期疾病患者,免疫治疗是一种有希望且有效的治疗方式。
{"title":"[Clinicopathological Analysis of 14 Cases of Primary Pulmonary Lymphoepithelial Carcinoma].","authors":"Yixuan Fang, Anzhe Wang, Lumin Shen, Xiao Yuan, Yu Kong","doi":"10.3779/j.issn.1009-3419.2024.101.29","DOIUrl":"10.3779/j.issn.1009-3419.2024.101.29","url":null,"abstract":"<p><strong>Background: </strong>Primary pulmonary lymphoepithelial carcinoma (PPLEC) is a rare form of lung malignancy, accounting for only 0.7% of all lung cancers. It is currently classified as a distinct subtype within squamous cell carcinomas. This study aims to explore the clinicopathological characteristics of PPLEC and its subtypes, with the objective of enhancing understanding and improving diagnostic accuracy for this disease.</p><p><strong>Methods: </strong>A retrospective analysis was conducted on the clinical, pathological, imaging, and prognostic data of 14 patients diagnosed with PPLEC at the First Affiliated Hospital of Soochow University between February 2019 and June 2023.</p><p><strong>Results: </strong>A total of 14 cases of PPLEC were identified, including 5 cases of the Regaud type, with ages ranging from 33 to 73 years, comprising 2 males and 3 females; and 9 cases of the Schmincke type, with ages ranging from 36 to 79 years, including 4 males and 5 females. Computed tomography (CT) scans consistently demonstrated soft tissue masses or nodular shadows. Reagud type mainly showed peripheral masses and Schmincke type mainly showed central masses. Pathological examination revealed tumor cells exhibiting syncytial-like growth, accompanied by lymphocytic infiltration and stromal fibrosis, with the Regaud type showing well-defined borders combined with granulomatous inflammation, while the Schmincke type exhibited indistinct tumor margins. Immunohistochemistry showed that CK, CK5/6, P40 and P63 were positive, and the Ki-67 index of Regaud type was lower than that of Schmincke type; notably, all 8 cases tested for programmed death-ligand 1 (PD-L1) were positive. Epstein-Barr virus-encoded RNA (EBER) in situ hybridization was positive in all instances. Among these cases, 6 underwent surgical treatment, and 8 received comprehensive therapy; by the end of the follow-up period, all 14 patients remained alive.</p><p><strong>Conclusions: </strong>PPLEC is a rare form of malignant lung tumor associated with Epstein-Barr virus (EBV) infection. The Regaud and Schmincke subtypes display distinct imaging and pathological characteristics. In the early stages of the disease, surgical intervention is the primary treatment method; however, for advanced stages, a multimodal treatment approach is utilized, resulting in a relatively favorable prognosis. Immunotherapy represents a promising and effective treatment modality for patients with middle to advanced stage disease exhibiting high PD-L1 expression levels.</p>","PeriodicalId":39317,"journal":{"name":"中国肺癌杂志","volume":"27 11","pages":"840-848"},"PeriodicalIF":0.0,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732381/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972557","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
[Mechanism of Bone-metastatic LUAD Cells Promoting Angiogenesis 
Through HGF/YAP Signaling Pathway]. [骨转移性LUAD细胞促进血管生成的机制
通过HGF/YAP信号通路]。
Q4 Medicine Pub Date : 2024-11-20 DOI: 10.3779/j.issn.1009-3419.2024.102.38
Yan Deng, Rong Qiu, Xingyu Liu, Yingyang Su, Yang Xue, Yuzhen Du

Background: The early stages of tumor bone metastasis are closely associated with changes in the vascular niche of the bone microenvironment, and abnormal angiogenesis accelerates tumor metastasis and progression. However, the effects of lung adenocarcinoma (LUAD) cells reprogrammed by the bone microenvironment on the vascular niche within the bone microenvironment and the underlying mechanisms remain unclear. This study investigates the effects and mechanisms of LUAD cells reprogrammed by the bone microenvironment on endothelial cells and angiogenesis, providing insights into the influence of tumor cells on the vascular niche within the bone microenvironment.

Methods: The culture media from bone-metastatic LUAD cell A549-GFP-LUC-BM3 (BM3-CM) and A549-GFP-LUC (A549-CM) were separately applied to human umbilical vein endothelial cell (HUVEC). A colony formation assay, scratch assay, and tube formation assay were conducted to evaluate the proliferation, migration, and angiogenesis of HUVEC. Gene set enrichment analysis (GSEA) was conducted to identify enriched pathways, while reverse transcription quantitative polymerase chain reaction (RT-qPCR) and enzyme linked immunosorbent assay (ELISA) were performed to quantify hepatocyte growth factor (HGF), a protein that plays a crucial role in angiogenesis. Furthermore, the pivotal function of HGF and its underlying molecular mechanisms have been substantiated through the utilisation of recombinant proteins, neutralising antibodies, pathway inhibitors, immunofluorescence staining, and Western blot.

Results: BM3-CM demonstrated a more pronounced impact on the proliferation, migration, and angiogenesis of HUVEC compared to A549-CM. Bioinformatics analysis, combined with in vitro experiment, demonstrated that the secretory protein HGF was significantly elevated in BM3 cells and BM3-CM (P<0.05). The addition of HGF neutralizing antibodies to BM3-CM inhibited the promoting effect of BM3-CM on HUVEC (P<0.05), while the addition of recombinant HGF to A549-CM reproduced that promoting effect of BM3-CM on HUVEC (P<0.05). HGF can enhance the activation of YAP (Yes-associated protein) in HUVEC, and this promotion effect may be achieved by activating Src and activating YAP into the nucleus (P<0.05), but this effect can be inhibited by HGF neutralizing antibodies (P<0.05). Furthermore, the addition of recombinant HGF to A549-CM can recapitulate the YAP activation effect of BM3-CM in HUVEC (P<0.05).

Conclusions: Bone microenvironment reprogrammed bone-metastatic LUAD cells BM3 promote the proliferation, migration, and angiogenesis of HUVEC through the HGF/YAP axis, potentially playing a significant role in the modifications of the vascular niche.

背景:肿瘤骨转移的早期阶段与骨微环境血管生态位的改变密切相关,异常的血管生成加速了肿瘤的转移和进展。然而,骨微环境重编程肺腺癌(LUAD)细胞对骨微环境内血管生态位的影响及其潜在机制尚不清楚。本研究探讨了骨微环境重编程LUAD细胞对内皮细胞和血管生成的影响及其机制,为肿瘤细胞对骨微环境内血管生态位的影响提供了新的思路。方法:将骨转移LUAD细胞A549-GFP-LUC- bm3 (BM3-CM)和A549-GFP-LUC (A549-CM)培养基分别应用于人脐静脉内皮细胞(HUVEC)。采用菌落形成实验、划痕实验和试管形成实验来评估HUVEC的增殖、迁移和血管生成。通过基因集富集分析(GSEA)确定富集途径,通过逆转录定量聚合酶链反应(RT-qPCR)和酶联免疫吸附试验(ELISA)定量肝细胞生长因子(HGF),这是一种在血管生成中起关键作用的蛋白质。此外,HGF的关键功能及其潜在的分子机制已经通过重组蛋白、中和抗体、途径抑制剂、免疫荧光染色和Western blot得到证实。结果:与A549-CM相比,BM3-CM对HUVEC的增殖、迁移和血管生成的影响更为明显。生物信息学分析结合体外实验证实BM3细胞和BM3- cm分泌蛋白HGF显著升高(p)。结论:骨微环境重编程骨转移LUAD细胞BM3通过HGF/YAP轴促进HUVEC的增殖、迁移和血管生成,可能在血管生态位的改变中发挥重要作用。
{"title":"[Mechanism of Bone-metastatic LUAD Cells Promoting Angiogenesis \u2029Through HGF/YAP Signaling Pathway].","authors":"Yan Deng, Rong Qiu, Xingyu Liu, Yingyang Su, Yang Xue, Yuzhen Du","doi":"10.3779/j.issn.1009-3419.2024.102.38","DOIUrl":"10.3779/j.issn.1009-3419.2024.102.38","url":null,"abstract":"<p><strong>Background: </strong>The early stages of tumor bone metastasis are closely associated with changes in the vascular niche of the bone microenvironment, and abnormal angiogenesis accelerates tumor metastasis and progression. However, the effects of lung adenocarcinoma (LUAD) cells reprogrammed by the bone microenvironment on the vascular niche within the bone microenvironment and the underlying mechanisms remain unclear. This study investigates the effects and mechanisms of LUAD cells reprogrammed by the bone microenvironment on endothelial cells and angiogenesis, providing insights into the influence of tumor cells on the vascular niche within the bone microenvironment.</p><p><strong>Methods: </strong>The culture media from bone-metastatic LUAD cell A549-GFP-LUC-BM3 (BM3-CM) and A549-GFP-LUC (A549-CM) were separately applied to human umbilical vein endothelial cell (HUVEC). A colony formation assay, scratch assay, and tube formation assay were conducted to evaluate the proliferation, migration, and angiogenesis of HUVEC. Gene set enrichment analysis (GSEA) was conducted to identify enriched pathways, while reverse transcription quantitative polymerase chain reaction (RT-qPCR) and enzyme linked immunosorbent assay (ELISA) were performed to quantify hepatocyte growth factor (HGF), a protein that plays a crucial role in angiogenesis. Furthermore, the pivotal function of HGF and its underlying molecular mechanisms have been substantiated through the utilisation of recombinant proteins, neutralising antibodies, pathway inhibitors, immunofluorescence staining, and Western blot.</p><p><strong>Results: </strong>BM3-CM demonstrated a more pronounced impact on the proliferation, migration, and angiogenesis of HUVEC compared to A549-CM. Bioinformatics analysis, combined with in vitro experiment, demonstrated that the secretory protein HGF was significantly elevated in BM3 cells and BM3-CM (P<0.05). The addition of HGF neutralizing antibodies to BM3-CM inhibited the promoting effect of BM3-CM on HUVEC (P<0.05), while the addition of recombinant HGF to A549-CM reproduced that promoting effect of BM3-CM on HUVEC (P<0.05). HGF can enhance the activation of YAP (Yes-associated protein) in HUVEC, and this promotion effect may be achieved by activating Src and activating YAP into the nucleus (P<0.05), but this effect can be inhibited by HGF neutralizing antibodies (P<0.05). Furthermore, the addition of recombinant HGF to A549-CM can recapitulate the YAP activation effect of BM3-CM in HUVEC (P<0.05).</p><p><strong>Conclusions: </strong>Bone microenvironment reprogrammed bone-metastatic LUAD cells BM3 promote the proliferation, migration, and angiogenesis of HUVEC through the HGF/YAP axis, potentially playing a significant role in the modifications of the vascular niche.</p>","PeriodicalId":39317,"journal":{"name":"中国肺癌杂志","volume":"27 11","pages":"805-814"},"PeriodicalIF":0.0,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732382/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972562","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
[Prognostic Value of STMN1 Expression in Non-small Cell Lung Cancer: 
A Meta-analysis]. [STMN1表达在非小细胞肺癌中的预后价值:
meta分析]。
Q4 Medicine Pub Date : 2024-11-20 DOI: 10.3779/j.issn.1009-3419.2024.102.39
Mengjie Li, Qinghua Zhou

Background: Lung cancer is one of the malignant tumors with the highest morbidity and mortality rates worldwide, seriously threatening human health. Non-small cell lung cancer (NSCLC) accounts for more than 85% of all lung cancer cases. STMN1 is a microtubule depolymerizing protein widely present in the cytoplasm and its expression level is associated with the prognosis of NSCLC patients. Through meta-analysis, this study aimed to investigate the predictive value of the expression level of STMN1 for the prognosis of lung cancer and screen for tumor markers with high sensitivity and specificity to optimize the whole-process management of lung cancer patients.

Methods: The PubMed, The Cochrane Library, Embase, WanFang and CNKI databases were searched from the inception to Sep 6, 2024 for relevant literature. The quality of included studies was assessed by the Newcastle-Ottawa Scale (NOS) score. The hazard ratio (HR) with 95%CI was combined to assess the relationship between STMN1 expression and prognostic factors. The prognostic indicators included the overall survival (OS) and disease-free survival (DFS). All statistical analysis was conducted by the STATA 17.0 software.

Results: A total of 5 high-quality studies (NOS score≥6 points) involving 754 patients were enrolled. The pooled results demonstrated that overexpression of STMN1 was significantly related to worse OS (HR=2.28, 95%CI: 1.79-2.91, P<0.001) and DFS (HR=2.14, 95%CI: 1.45-3.17, P<0.001). Overexpression of STMN1 was a risk factor for poor prognosis of NSCLC patients.

Conclusions: Overexpression of STMN1 is a poor prognostic factor in NSCLC patients. STMN1 may serve as a prognostic biomarker for NSCLC patients. However, more researches are still needed to verify the above findings.

背景:肺癌是世界上发病率和死亡率最高的恶性肿瘤之一,严重威胁着人类的健康。非小细胞肺癌(NSCLC)占所有肺癌病例的85%以上。STMN1是一种广泛存在于细胞质中的微管解聚蛋白,其表达水平与NSCLC患者的预后相关。本研究旨在通过meta分析,探讨STMN1表达水平对肺癌预后的预测价值,筛选敏感性和特异性较高的肿瘤标志物,优化肺癌患者的全过程管理。方法:检索PubMed、Cochrane Library、Embase、万方、CNKI等数据库自建站至2024年9月6日的相关文献。纳入研究的质量采用纽卡斯尔-渥太华量表(NOS)评分进行评估。合并95%CI的风险比(HR)来评估STMN1表达与预后因素的关系。预后指标包括总生存期(OS)和无病生存期(DFS)。所有统计分析均采用STATA 17.0软件进行。结果:共纳入5项高质量研究(NOS评分≥6分),共纳入754例患者。综合结果显示,STMN1过表达与较差的OS有显著相关性(HR=2.28, 95%CI: 1.79 ~ 2.91)。结论:STMN1过表达是NSCLC患者预后不良的因素。STMN1可作为非小细胞肺癌患者的预后生物标志物。然而,上述发现还需要更多的研究来验证。
{"title":"[Prognostic Value of STMN1 Expression in Non-small Cell Lung Cancer: \u2029A Meta-analysis].","authors":"Mengjie Li, Qinghua Zhou","doi":"10.3779/j.issn.1009-3419.2024.102.39","DOIUrl":"10.3779/j.issn.1009-3419.2024.102.39","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer is one of the malignant tumors with the highest morbidity and mortality rates worldwide, seriously threatening human health. Non-small cell lung cancer (NSCLC) accounts for more than 85% of all lung cancer cases. STMN1 is a microtubule depolymerizing protein widely present in the cytoplasm and its expression level is associated with the prognosis of NSCLC patients. Through meta-analysis, this study aimed to investigate the predictive value of the expression level of STMN1 for the prognosis of lung cancer and screen for tumor markers with high sensitivity and specificity to optimize the whole-process management of lung cancer patients.</p><p><strong>Methods: </strong>The PubMed, The Cochrane Library, Embase, WanFang and CNKI databases were searched from the inception to Sep 6, 2024 for relevant literature. The quality of included studies was assessed by the Newcastle-Ottawa Scale (NOS) score. The hazard ratio (HR) with 95%CI was combined to assess the relationship between STMN1 expression and prognostic factors. The prognostic indicators included the overall survival (OS) and disease-free survival (DFS). All statistical analysis was conducted by the STATA 17.0 software.</p><p><strong>Results: </strong>A total of 5 high-quality studies (NOS score≥6 points) involving 754 patients were enrolled. The pooled results demonstrated that overexpression of STMN1 was significantly related to worse OS (HR=2.28, 95%CI: 1.79-2.91, P<0.001) and DFS (HR=2.14, 95%CI: 1.45-3.17, P<0.001). Overexpression of STMN1 was a risk factor for poor prognosis of NSCLC patients.</p><p><strong>Conclusions: </strong>Overexpression of STMN1 is a poor prognostic factor in NSCLC patients. STMN1 may serve as a prognostic biomarker for NSCLC patients. However, more researches are still needed to verify the above findings.</p>","PeriodicalId":39317,"journal":{"name":"中国肺癌杂志","volume":"27 11","pages":"826-830"},"PeriodicalIF":0.0,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732388/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972572","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
[Radiotherapy Combined with Immunotherapy and Chemotherapy Improves Prognosis and Demonstrates Synergistic Effects in Extensive-stage Small Cell Lung Cancer]. 【放疗联合免疫化疗改善广泛期小细胞肺癌的预后并显示协同作用】。
Q4 Medicine Pub Date : 2024-11-20 DOI: 10.3779/j.issn.1009-3419.2024.102.41
Huaijun Ji, Meiling Sun, Jingyi Li, Ge Yu, Yongbing Chen

Background: Extensive-stage small cell lung cancer (ES-SCLC) is a malignant tumor with remarkable proliferative and invasive ability, which has very poor clinical prognosis due to lack of effective treatments. This study aims to evaluate the efficacy and synergistic effects of radiotherapy (RT) combined with immunotherapy (IT) and chemotherapy (CT) in patients with ES-SCLC.

Methods: A retrospective analysis was performed on 145 ES-SCLC patients treated with first-line CT. Kaplan-Meier analysis and Log-rank tests were used to evaluate survival outcomes, while propensity score matching (PSM) was applied to reduce confounding factors.

Results: The median overall survival (mOS) and median progression-free survival (mPFS) for the entire cohort were 15.7 and 6.9 mon, respectively. The IT+CT group had a significantly longer mOS compared to the CT group (17.2 vs 13.5 mon, P=0.047). Similarly, the RT+CT group demonstrated superior mOS (18.5 vs 12.3 mon, P<0.001) and mPFS (7.1 vs 6.2 mon, P=0.006) compared to the CT group. Multivariate analysis identified RT, IT, and Eastern Cooperative Oncology Group performance status (ECOG PS) as independent prognostic factors for mOS (P<0.05), while gender and ECOG PS were independent predictors for mPFS (P<0.05). Following PSM, the RT+CT group continued to exhibit significant advantages in mOS (18.0 vs 12.1 mon, P<0.001) and mPFS (7.1 vs 5.5 mon, P=0.037) compared to the CT group. Notably, the RT+IT+CT group achieved a markedly longer mOS than the IT+CT group (28.5 vs 15.8 mon, P=0.017). Grade 3-4 adverse events occurred in 27.6% of patients, with no grade 5 adverse events reported.

Conclusions: The combination of RT, IT, and CT significantly enhances the prognosis of ES-SCLC patients. RT plays a key role in their synergistic effects and demonstrates good safety, warranting further research and clinical application.

背景:广泛期小细胞肺癌(ES-SCLC)是一种具有显著增生性和侵袭性的恶性肿瘤,由于缺乏有效的治疗,临床预后非常差。本研究旨在评价放疗(RT)联合免疫治疗(IT)和化疗(CT)治疗ES-SCLC的疗效及协同效应。方法:回顾性分析145例ES-SCLC患者行一线CT治疗的临床资料。Kaplan-Meier分析和Log-rank检验用于评估生存结果,倾向评分匹配(PSM)用于减少混杂因素。结果:整个队列的中位总生存期(mOS)和中位无进展生存期(mPFS)分别为15.7和6.9个月。与CT组相比,IT+CT组的mOS明显更长(17.2个月vs 13.5个月,P=0.047)。同样,RT+CT组表现出更高的mOS(18.5月vs 12.3月)。结论:RT、IT和CT联合治疗可显著改善ES-SCLC患者的预后。RT在其协同作用中起关键作用,且具有良好的安全性,值得进一步研究和临床应用。
{"title":"[Radiotherapy Combined with Immunotherapy and Chemotherapy Improves Prognosis and Demonstrates Synergistic Effects in Extensive-stage Small Cell Lung Cancer].","authors":"Huaijun Ji, Meiling Sun, Jingyi Li, Ge Yu, Yongbing Chen","doi":"10.3779/j.issn.1009-3419.2024.102.41","DOIUrl":"10.3779/j.issn.1009-3419.2024.102.41","url":null,"abstract":"<p><strong>Background: </strong>Extensive-stage small cell lung cancer (ES-SCLC) is a malignant tumor with remarkable proliferative and invasive ability, which has very poor clinical prognosis due to lack of effective treatments. This study aims to evaluate the efficacy and synergistic effects of radiotherapy (RT) combined with immunotherapy (IT) and chemotherapy (CT) in patients with ES-SCLC.</p><p><strong>Methods: </strong>A retrospective analysis was performed on 145 ES-SCLC patients treated with first-line CT. Kaplan-Meier analysis and Log-rank tests were used to evaluate survival outcomes, while propensity score matching (PSM) was applied to reduce confounding factors.</p><p><strong>Results: </strong>The median overall survival (mOS) and median progression-free survival (mPFS) for the entire cohort were 15.7 and 6.9 mon, respectively. The IT+CT group had a significantly longer mOS compared to the CT group (17.2 vs 13.5 mon, P=0.047). Similarly, the RT+CT group demonstrated superior mOS (18.5 vs 12.3 mon, P<0.001) and mPFS (7.1 vs 6.2 mon, P=0.006) compared to the CT group. Multivariate analysis identified RT, IT, and Eastern Cooperative Oncology Group performance status (ECOG PS) as independent prognostic factors for mOS (P<0.05), while gender and ECOG PS were independent predictors for mPFS (P<0.05). Following PSM, the RT+CT group continued to exhibit significant advantages in mOS (18.0 vs 12.1 mon, P<0.001) and mPFS (7.1 vs 5.5 mon, P=0.037) compared to the CT group. Notably, the RT+IT+CT group achieved a markedly longer mOS than the IT+CT group (28.5 vs 15.8 mon, P=0.017). Grade 3-4 adverse events occurred in 27.6% of patients, with no grade 5 adverse events reported.</p><p><strong>Conclusions: </strong>The combination of RT, IT, and CT significantly enhances the prognosis of ES-SCLC patients. RT plays a key role in their synergistic effects and demonstrates good safety, warranting further research and clinical application.</p>","PeriodicalId":39317,"journal":{"name":"中国肺癌杂志","volume":"27 11","pages":"831-839"},"PeriodicalIF":0.0,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732380/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972577","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
[Advances of Neoadjuvant Targeted Therapy in ALK-positive Non-small Cell Lung Cancer]. [alk阳性非小细胞肺癌新辅助靶向治疗进展]。
Q4 Medicine Pub Date : 2024-11-20 DOI: 10.3779/j.issn.1009-3419.2024.106.30
Weizhen Sun, Yuheng Zhou, Yaobin Lin, Shoucheng Feng, Hao Long

Lung cancer remains the most frequently diagnosed cancer and the leading cause of cancer-related death worldwide, with anaplastic lymphoma kinase (ALK) fusion mutations accounting for approximately 4%-9% of cases. In recent years, there are increasing clinical evidences suggesting that the combination of ALK inhibitors with surgical treatment holds significant potential for clinical application in resectable early and locally advanced non-small cell lung cancer (NSCLC) patients. This review aims to summarize the advances in neoadjuvant targeted therapy for ALK fusion positive NSCLC and discuss its advantages and challenges in clinical practice.
.

肺癌仍然是世界范围内最常见的癌症和癌症相关死亡的主要原因,间变性淋巴瘤激酶(ALK)融合突变约占病例的4%-9%。近年来,越来越多的临床证据表明,ALK抑制剂联合手术治疗在可切除的早期和局部晚期非小细胞肺癌(NSCLC)患者中具有重要的临床应用潜力。本文旨在总结ALK融合阳性NSCLC新辅助靶向治疗的进展,并讨论其在临床实践中的优势和挑战。
。
{"title":"[Advances of Neoadjuvant Targeted Therapy in ALK-positive Non-small Cell Lung Cancer].","authors":"Weizhen Sun, Yuheng Zhou, Yaobin Lin, Shoucheng Feng, Hao Long","doi":"10.3779/j.issn.1009-3419.2024.106.30","DOIUrl":"10.3779/j.issn.1009-3419.2024.106.30","url":null,"abstract":"<p><p>Lung cancer remains the most frequently diagnosed cancer and the leading cause of cancer-related death worldwide, with anaplastic lymphoma kinase (ALK) fusion mutations accounting for approximately 4%-9% of cases. In recent years, there are increasing clinical evidences suggesting that the combination of ALK inhibitors with surgical treatment holds significant potential for clinical application in resectable early and locally advanced non-small cell lung cancer (NSCLC) patients. This review aims to summarize the advances in neoadjuvant targeted therapy for ALK fusion positive NSCLC and discuss its advantages and challenges in clinical practice.\u2029.</p>","PeriodicalId":39317,"journal":{"name":"中国肺癌杂志","volume":"27 11","pages":"849-854"},"PeriodicalIF":0.0,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732383/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142972553","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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
中国肺癌杂志
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